1
|
Chou MY, Yang MH. Immunomodulation on tumor immune microenvironment in acquired targeted therapy resistance and implication for immunotherapy resistance. Transl Oncol 2025; 54:102353. [PMID: 40058234 PMCID: PMC11929932 DOI: 10.1016/j.tranon.2025.102353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025] Open
Abstract
The emergence of molecularly targeted therapies and immunotherapies has revolutionized cancer treatment, yet the optimal sequencing of these modalities remains debated. While targeted therapies often induce initial immunostimulatory effects, the development of resistance is accompanied by dynamic alterations in the tumor-immune microenvironment. These changes can promote tumor growth, hinder immune surveillance, and contribute to subsequent immunotherapy resistance. This review focuses on solid tumors and summarizes the immunomodulatory effects arising in the context of targeted therapy resistance, highlighting the challenges they pose for the subsequent immunotherapy efficacy.
Collapse
Affiliation(s)
- Ming-Yu Chou
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei 112304, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan.
| |
Collapse
|
2
|
Huang T, Huang W, Huang G, Wei X, Huang Y, Liu T, Liu Y, Ni W, Che C. Pincer-Type Pt(II)-NHC Antibody-Drug Conjugate for HER-2-Targeted Chemoimmunotherapy. Adv Healthc Mater 2025; 14:e2403449. [PMID: 39950551 PMCID: PMC11973945 DOI: 10.1002/adhm.202403449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/28/2025] [Indexed: 04/08/2025]
Abstract
Platinum-based chemotherapy drugs play an indispensable role in clinical cancer treatment, but exhibit considerable side effects due to their non-specific mechanism of killing cancer cells and normal cells. In this regard, the use of antibodies conjugated to anti-cancer platinum complexes will enable better differentiation of cancer cells from normal cells. Here, six pincer-platinum(II) NHC (N-heterocyclic carbene) complexes are reported, one of which has an amino group on the N-alkyl group of the NHC ligand. This platinum(II) complex is used as the payload for platinum(II)-based antibody-drug conjugate (ADC) targeting human epidermal growth factor receptor 2 (HER-2). Notably, this ADC can specifically bind to the HER-2 antigen, distinguish target cells from non-target cells, and exhibit good anti-tumor activity in vitro and in vivo.
Collapse
Affiliation(s)
- Tao Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Wan‐Qiong Huang
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Gui‐Feng Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Xiao‐Long Wei
- Department of PathologyCancer Hospital of Shantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Yong‐Liang Huang
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
| | - Tao Liu
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Yungen Liu
- Department of ChemistrySouthern University of Science and TechnologyShenzhenGuangdong518055P. R. China
| | - Wen‐Xiu Ni
- Department of Medicinal ChemistryShantou University Medical CollegeShantouGuangdong515041P. R. China
- Chemistry and Chemical Engineering Guangdong LaboratoryShantouGuangdong515021P. R. China
| | - Chi‐Ming Che
- State Key Laboratory of Synthetic Chemistry and Department of ChemistryThe University of Hong KongHong KongP. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology LimitedHong Kong Science and Technology ParksUnits 1503–1511, 15/F, Building 17W, New TerritoriesHong KongP. R. China
| |
Collapse
|
3
|
Ruan X, Wu L, Tang Z, Li Y, Wang J, Jiang H, Zhang L, Wang S, Chen Z, Yuan C, Xia Y, Pan Y, Gao J, Zhao X. Two chemotherapeutic agents expand stem-like CD62L +CD8 + T cells in antitumor immune responses. Front Immunol 2025; 16:1533857. [PMID: 40236705 PMCID: PMC11996895 DOI: 10.3389/fimmu.2025.1533857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction Recent findings reveal that the precursors of exhausted CD8+ T (CD8+ Tpex) cells possess stem-like signatures in tumor immunity, which originate from tumor draining lymph node (TdLN)-derived tumor-specific memory (CD8+ TTSM) cells. Both of these T subsets can be collectively referred to as stem-like CD8+ T cells, which demonstrate robust self-renewal ability and can proliferate and differentiate into transitory effector-like exhausted T cells (Texint). There are reports that chemotherapeutic drugs can promote the antitumor immune responses of patients by increasing the number of CD8+ T cells; however, whether chemotherapeutic drugs increase these two stem-like CD8+ T cells remain further exploration. Methods Tpex cell-associated subpopulations in human colorectal tumors were analyzed by using single-cell sequencing data. CT26 and B16 tumor models of wild type and Eomes conditional knockout mice were constructed, and the changes of TTSM, Tpex and Tex subsets in mice were dissected by flow cytometry after treatment with decitabine (DAC), doxorubicin (DOX) and 5-Fluorouracil (5-FU). Results In this study, we demonstrated that DAC and 5-FU expanded CD8+ TTSM cells in TdLNs. At the same time, we validated that DAC and 5-FU substantially promoted the expansion of CD62L+CD8+ Tpex cells and subsequently increased effector function of CX3CR1+ CD8+ Texint cells. In addition, the conditional knockout of transcription factor Eomes in CD8+ T cells partially eliminated DAC-amplified CD62L+ CD8+ Tpex cells, but had no effect on such CD8+ T subset expanded by 5-FU. Conclusion The present study demonstrated that both DAC and 5-FU promoted the differentiation of stem-like CD8+ TTSM cells in TdLNs and significantly enhanced the differentiation and expansion of stem-like CD62L+ CD8+ Tpex and CX3CR1+ Texint cells in tumor microenvironment. The knockout of Eomes partially influenced the role of DAC in promoting the differentiation and expansion of stem-like CD8+ T cells.
Collapse
Affiliation(s)
- Xiaokang Ruan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of General Surgery, People's Hospital of Dongxihu District, Wuhan, China
| | - Linwei Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zijian Tang
- The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Haolin Jiang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengjia Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhaoqiang Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenlei Yuan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujian Xia
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Pan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianling Gao
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Simon N, Chandran E, Atiq S, Kydd AR, Girardi D, Ley L, Cordes L, Wang TF, Boudjadi S, Stukes I, Smith E, Akbulut D, Niglio S, Patel R, Banday R, Redd B, Gurram S, Steinberg S, Apolo AB. A phase II study of lurbinectedin with or without avelumab in small cell carcinoma of the bladder (laser)-design and rationale. Future Oncol 2025; 21:1171-1177. [PMID: 40116623 PMCID: PMC11988252 DOI: 10.1080/14796694.2025.2480534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Small cell carcinoma of the bladder is a rare, aggressive malignancy accounting for less than 1% of all bladder malignancies. Treatment regimens are drawn from the small cell lung cancer (SCLC) literature, with platinum and etoposide commonly used in the first-line setting. Unfortunately, responses are generally short-lived, and most patients relapse. There is little evidence to guide selection of later lines of therapy. Lurbinectedin is an alkylating agent with accelerated US FDA approval for use in patients with SCLC. Immune checkpoint inhibitors have also been approved for SCLC, improving survival when added to chemotherapy. This article describes the design and rationale behind LASER, an open-label phase II trial of lurbinectedin with or without avelumab.Clinical trial registration: NCT06228066 (ClinicalTrial.gov).
Collapse
Affiliation(s)
- Nicholas Simon
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elias Chandran
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Saad Atiq
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andre R. Kydd
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Girardi
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Ley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tzu-Fang Wang
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Salah Boudjadi
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ian Stukes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth Smith
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dilara Akbulut
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Scot Niglio
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Ruchi Patel
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rouf Banday
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bernadette Redd
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seth Steinberg
- Office of Collaborative Biostatistics, Office of the Clinical Director, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Wang Y, Zhou H, Ju S, Dong X, Zheng C. The solid tumor microenvironment and related targeting strategies: a concise review. Front Immunol 2025; 16:1563858. [PMID: 40207238 PMCID: PMC11979131 DOI: 10.3389/fimmu.2025.1563858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
The malignant tumor is a serious disease threatening human life. Increasing studies have confirmed that the tumor microenvironment (TME) is composed of a variety of complex components that precisely regulate the interaction of tumor cells with other components, allowing tumor cells to continue to proliferate, resist apoptosis, evade immune surveillance and clearance, and metastasis. However, the characteristics of each component and their interrelationships remain to be deeply understood. To target TME, it is necessary to deeply understand the role of various components of TME in tumor growth and search for potential therapeutic targets. Herein, we innovatively classify the TME into physical microenvironment (such as oxygen, pH, etc.), mechanical microenvironment (such as extracellular matrix, blood vessels, etc.), metabolic microenvironment (such as glucose, lipids, etc.), inflammatory microenvironment and immune microenvironment. We introduce a concise but comprehensive classification of the TME; depict the characteristics of each component in TME; summarize the existing methods for detecting each component in TME; highlight the current strategies and potential therapeutic targets for TME; discuss current challenges in presenting TME and its clinical applications; and provide our prospect on the future research direction and clinical benefits of TME.
Collapse
Affiliation(s)
- Yingliang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Huimin Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuguang Ju
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| |
Collapse
|
6
|
Deng Q, Wu L, He J, Wu F, Jiang Z. Identification of autophagy-related immune targets for enhancing immunotherapy in pancreatic cancer aggressiveness. Discov Oncol 2025; 16:382. [PMID: 40126694 PMCID: PMC11933596 DOI: 10.1007/s12672-025-02190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) presents significant challenges in oncology, with metastasis critically affecting patient outcomes. Autophagy-related genes (ARGs)'s involvement in influencing immune activity and metastasis in PC remains inadequately understood. AIM This study seeks to identify and validate five ARGs that could serve as immune targets, enhancing enhancing Pancreatic cancer metastasis (PCM)'s prognostic models and informing immunotherapy strategies. METHODS ARGs that were diffentially expressed were screened, followed by Cox regression and LASSO analyses to pinpoint five genes linked to overall survival (OS). A prognostic model was developed and validated using ROC curves. Functional analyses, including GO and KEGG, were performed to elucidate ARG mechanisms. Immune infiltration and TFs/microRNA/mRNA networks were assessed to understand ARG-immune cell interactions. Experimental validation employed real-time PCR, IHC, and Western blotting, supported by TCGA data. Functional assays explored RHEB's role in PC, particularly its interaction with LC3. RESULTS Five ARGs (CASP1, RHEB, CHMP2B, MYC, and HDAC6) were identified, contributing to a robust prognostic model where low-risk individuals showed significantly longer OS. The model demonstrated high AUC scores, indicating strong prognostic capability. CD8 T cells and Treg cells' elevated levels were observed in metastatic subjects. RHEB knockdown suppressed cancer cell proliferation and invasion, with a negative correlation between RHEB and LC3, suggesting a role in autophagy-mediated modulation of PC metastasis. CONCLUSION This study introduces a novel prognostic model incorporating five ARGs, highlighting their potential as immune targets for cancer immunotherapy. The negative correlation between RHEB and LC3 suggests a therapeutic pathway for PCM intervention, laying the groundwork for more effective anti-cancer strategies. These findings advance the identification of novel immune targets and signaling pathways, aligning with precision medicine goals in cancer treatment.
Collapse
Affiliation(s)
- Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
- Department of Gastroenterology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Linju Wu
- Department of Anesthesiology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 2621000, Sichuan, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
7
|
Zhao L, Zhang Z, Wang D, Yang L, Liu Z, Lou C. Analysis of the effectiveness and safety of lenvatinib/bevacizumab combined with PD-1/PD-L1 inhibitors and GEMOX in the first-line treatment of advanced biliary tract carcinoma. Clin Exp Med 2025; 25:87. [PMID: 40106138 PMCID: PMC11922973 DOI: 10.1007/s10238-025-01623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
To assess the efficacy and safety of lenvatinib/bevacizumab combined with programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors and gemcitabine/oxaliplatin (GEMOX) as first-line treatments in patients with advanced biliary tract cancer (BTC). Patients with advanced BTC who received lenvatinib/bevacizumab combined with PD-1/PD-L1 inhibitors plus gemcitabine/oxaliplatin (GEMOX) chemotherapy were retrospectively screened. The primary endpoints were overall survival (OS) and progression-free survival (PFS), whereas the secondary endpoints were objective response rate (ORR), disease control rate (DCR), and safety. Prognostic factors for survival were analyzed. A total of 172 individuals were enrolled and categorized into four groups: Group A received GEMOX plus PD-1 antibody (sintilimab or camrelizumab) and lenvatinib; group B received GEMOX and PD-1 antibody (sintilimab or camrelizumab) and bevacizumab; group C received GEMOX and PD-1 antibody (sintilimab or camrelizumab); and group D received GEMOX alone. The median OS was 13.63 months (95% confidence interval [CI]: 12.37-14.89), 12.41 months (95% CI: 10.67-12.32), 11.23 months (95% CI: 9.39-13.07), and 8.86 months (95% CI: 7.28-10.44) in groups A, B, C, and D, respectively (P = 0.312). In groups A, B, C, and D, the median PFS was 12.42 months, 11.05 months, 8.89 months, and 6.02 months. A statistically significant difference was observed (t = 2, 95% CI: 11.31-13.53, P < 0.01). The ORR was 45.00% (17/40) in group A, 34.78% (16/46) in group B, 16.67% (5/30) in group C, and 17.86% (10/56) in group D. The DCR was 87.50% (35/40), 78.26% (36/46), 76.67% (23/30), and 58.93% (33/56) in groups A, B, C, and D, respectively. In addition, regression analysis showed that patients' metastasis site, whether the neutrophil-lymphocyte ratio was < 2.3, and whether chemotherapy was administered through hepatic artery embolization and was independent prognostic factors influencing median OS and PFS. Almost all patients included in the study experienced treatment-related adverse events (TRAEs) of varying degrees of severity, with grade 1-2 adverse events predominating. Lenvatinib/bevacizumab combined with programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors and gemcitabine/oxaliplatin (GEMOX) represent an effective and tolerable regimen for advanced BTC in a multicenter retrospective real-world study.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Zhengfeng Zhang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Dazhen Wang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Ze Liu
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Changjie Lou
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.
| |
Collapse
|
8
|
Iturri L, Jouglar E, Gilbert C, Espenon J, Juchaux M, Prezado Y. A first evaluation of the efficacy of minibeam radiation therapy combined with an immune check point inhibitor in a model of glioma-bearing rats. Clin Transl Radiat Oncol 2025; 51:100911. [PMID: 39898330 PMCID: PMC11783053 DOI: 10.1016/j.ctro.2025.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 02/04/2025] Open
Abstract
Glioblastoma multiforme (GBM) continues to be a hopeless case today. Its treatment involves the use of multiple modalities. One of them is radiation therapy (RT), that is limited by normal tissue tolerances in GBM patients. GBM is widely recognized to induce local and systemic immunosuppression, which is a hindrance to the use of immune-modulating therapies. One possible strategy is to ally immunotherapy (IT) with novel RT approaches able to revert the immunosuppressive nature of GBM. One example is minibeam radiation therapy (MBRT). Preclinical experiments have shown that MBRT leads to a remarkable widening of the therapeutic window for GBM and elicits an effective immune priming. The main hypothesis of this study is that the activation of the immune system by MBRT would synergize with IT enhancing tumour control and minimizing toxicities. To validate it, in vivo experiments in a glioma rat model were performed. The goal was to assess the gain in survival of animals treated with MBRT, MBRT plus an immune check point inhibitor (ICI) versus conventional RT (CRT) or ICI alone. All treatments (ICI alone, CRT, CRT + ICI, MBRT and MBRT + ICI) increased survival with respect to the non-irradiated controls. However, the high radiation dose (30 Gy) delivered in one fraction in CRT is highly detrimental for normal tissues contrary to MBRT. The combination of CRT plus ICI appeared to be toxic. MBRT + ICI surpassed the survival rate with respect to ICI alone. When ICI was used with high-dose conventional irradiation, tumor eradication was observed in 6 out 8 animals. However, the survival was statistically equivalent to MBRT plus ICI, with a tendency to reduce survival in comparison with CRT alone, suggesting acute toxicity associated with this multimodal treatment. In conclusion, our results suggest some advantages for MBRT in combination with ICI. We need to conduct further work to determine the optimal RT-IT combination and schedule for ICI injection and MBRT irradiation.
Collapse
Affiliation(s)
- Lorea Iturri
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Emmanuel Jouglar
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Institut Curie, PSL Research University, Department of Radiation Oncology - Paris and Orsay Protontherapy Centre, F-75005 Paris, France
| | - Cristèle Gilbert
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Julie Espenon
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Marjorie Juchaux
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- New Approaches in Radiotherapy Lab, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, A Coruña, Spain
- Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, A Coruña, Spain
| |
Collapse
|
9
|
Liu QQ, Yan J, Ye YF, Yang CN, Chen ZJ, Lin HM, Zhang ZT, Zhang R. Efficacy and conversion outcome of chemotherapy combined with PD-1 inhibitor for patients with unresectable or recurrent gallbladder carcinoma: a real-world exploratory study. World J Surg Oncol 2025; 23:69. [PMID: 40022061 PMCID: PMC11869664 DOI: 10.1186/s12957-025-03703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/03/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is an extremely aggressive tumor of the biliary tract with a bleak prognosis, and the evidence supporting the benefit of available systemic therapy for advanced GBC is scarce. Herein, this study intended to investigate the real-world outcome of chemotherapy combined with programmed death-1 (PD-1) inhibitor for the management of unresectable or recurrent GBC. METHODS From January 2018 to December 2023, consecutive patients who were treated with systematic treatment, including chemotherapy or the combination of chemotherapy plus PD-1 inhibitor, for unresectable or recurrent GBC were retrospectively identified. Clinical data regarding baseline characteristics, therapeutic response, adverse events (AEs), and oncological outcomes were collected. RESULTS The eligible patients were allocated to combination therapy arm (n = 46) and mono-chemotherapy arm (n = 19). After propensity score matching (PSM), 16 patients were allocated in each arm. The overall survival (OS) and progression-free survival (PFS) of combination therapy were marginally superior to mono-chemotherapy both before and after PSM. The combination therapy exhibited advantage over mono-chemotherapy in regards to partial response (PR) (before PSM: P = 0.009; after PSM: P = 0.037) and objective response rate (ORR) (before PSM: P = 0.006; after PSM: P = 0.015). In combined therapy cohort, 1 patient achieve a complete response, and 13 patients were assessed as appropriate for surgical excision, among which 1 patient refused further surgical intervention. CONCLUSIONS In patients with unresectable or recurrent GBC, the combination of chemotherapy and PD-1 inhibitor as first-line therapy exhibited prolonged OS and PFS, and increased PR and ORR over those receiving chemotherapy alone, with an acceptable toxicity profile. The combination therapy may be a potential conversion therapy in unresectable GBC patients.
Collapse
Affiliation(s)
- Qin-Qin Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Guangdong, China
| | - Jian Yan
- Department of Hepatobiliary surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Fang Ye
- Clinical Research Design Division, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Cai-Ni Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhi-Jun Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hao-Ming Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | | | - Rui Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| |
Collapse
|
10
|
Yi L, Gu Y, Guo L, Zou X, Wu G. Effect of chemoradiotherapy on the dynamics of circulating lymphocyte subsets in patients with non-metastatic nasopharyngeal carcinoma. Front Oncol 2025; 15:1521836. [PMID: 40012553 PMCID: PMC11861370 DOI: 10.3389/fonc.2025.1521836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Background Chemoradiotherapy (CRT) is the primary and most effective treatment for non-metastatic nasopharyngeal carcinoma (NPC), exerting antitumor effects by modulating immune cells. Distinct subpopulations of immune cells exhibit specific sensitivity to CRT. This study aimed to characterize the dynamics of the proportions and absolute counts of peripheral circulating lymphocyte subsets in non-metastatic NPC before and after CRT, and to elucidate their association with clinical responses. Methods A total of 91 patients with non-metastatic NPC were enrolled. Flow cytometry was employed to detect the expression of CD3, CD4, CD8, CD56, and CD19 on peripheral blood cells. The composition of lymphocyte subsets before treatment, post-completion of CRT, and one month following CRT was retrospectively analyzed. Further, the relationship between the composition of circulating lymphocyte subpopulations and distinguish clinical responses was evaluated. Results The proportion of CD3+ T cells showed an initial increase followed by a significant decrease at baseline, post-completion of CRT, and one month following CRT. The proportions of CD3+CD4+ T cells, CD4+/CD8+ ratio, and CD19+ B cells continued to decline at baseline, post-completion of CRT, and one month following CRT, while the proportions of CD3+CD8+ T cells and CD16+CD56+ NK cells progressively increased. The absolute counts of circulating lymphocyte subsets, including CD3+ T cells, CD3+CD4+ T cells, CD3+CD8+ T cells, CD45+, CD19+ B cells, and CD16+CD56+ NK cells, demonstrated a trend of initial decrease followed by an increase at baseline, post-completion of CRT, and one month following CRT. Patients with complete response (CR) and partial response (PR) presented similar dynamic trends in the percentages and absolute counts of circulating lymphocyte subpopulations at baseline, post-completion of CRT, and one month following CRT. The proportions and absolute counts of CD3+CD4+ T cells in CR patients were distinctly higher than those in PR patients at the end of CRT, whereas the absolute counts of CD16+CD56+ NK cells were remarkably lower in CR patients compared to PR patients. The baseline proportion and absolute count of CD19+ B cells, as well as the absolute count of CD3+CD4+ T cells, were significantly higher in CR patients compared with PR patients. Conclusion CRT induced dynamic alterations in the peripheral lymphocyte profile of non-metastatic NPC patients. Assessing the variations in the distribution of circulating lymphocyte subsets among patients with different clinical treatment responses will be helpful in developing protocols for the concurrent utilization of immunotherapeutic drugs and CRT.
Collapse
Affiliation(s)
- Lilan Yi
- Department of Oncology, Cancer Center, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, Guangdong, China
| | - Yinfang Gu
- Department of Oncology, Cancer Center, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, Guangdong, China
| | - Longhua Guo
- Department of Oncology, Cancer Center, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, Guangdong, China
| | - Xiaofang Zou
- Department of Oncology, Cancer Center, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, Guangdong, China
| | - Guowu Wu
- Department of Oncology, Cancer Center, Meizhou People’s Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, Guangdong, China
| |
Collapse
|
11
|
Zhang L, Shi J, Zhu MH, Huang Y, Lu Q, Sun P, Chen HZ, Lai X, Fang C. Liposomes-enabled cancer chemoimmunotherapy. Biomaterials 2025; 313:122801. [PMID: 39236630 DOI: 10.1016/j.biomaterials.2024.122801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.
Collapse
Affiliation(s)
- Lele Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiangpei Shi
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanhu Huang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
12
|
Bai Z, Yang Y, Cui Z, Liang W, Zhang X, Zhang Z, Sun J, Liu Z, Li K, Shi M, Li J. Double-targeted liposomes coated with matrix metallopeptidase-2-responsive polypeptide nanogel for chemotherapy and enhanced immunotherapy against cervical cancer. Mater Today Bio 2025; 30:101412. [PMID: 39811606 PMCID: PMC11731983 DOI: 10.1016/j.mtbio.2024.101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Immunotherapy is a cornerstone in cancer treatment, celebrated for its precision, ability to eliminate residual cancer cells, and potential to avert tumor recurrence. Nonetheless, its effectiveness is frequently undermined by the immunosuppressive milieu created by tumors. This study presents a novel nanogel-based drug delivery system, DOX-4PI@CpG@Lipo@Gel (DPCLG), engineered to respond to Matrix Metallopeptidase-2 (MMP-2)-a protease abundant in the tumor microenvironment (TME). This system enables the controlled release of two distinct types of liposomes within the TME. The first, DOX-4PI@Liposome (DPL), carries doxorubicin (DOX) and 4-phenylimidazole (4PI), targeting cancer cells to provide chemotherapeutic effects while diminishing the immunosuppressive environment. The second, a mannosyl-modified cationic liposome (CL), is loaded with Cytosine phosphate guanine (CpG) oligodeoxynucleotides to specifically target M2 phenotype macrophages, reversing their tumor-associated phenotype (TAM) and activating immune cytokines to promote tumor destruction. Our findings indicate that DPCLG significantly curtails tumor growth, both in vitro and in vivo, mitigates the immunosuppressive TME, and triggers a potent systemic immune response. This study underscores the potential of DPCLG as an advanced, dual-targeting drug delivery system for comprehensive cancer therapy.
Collapse
Affiliation(s)
- Zhimin Bai
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Yibo Yang
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Zutong Cui
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Wenming Liang
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Xin Zhang
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Zihan Zhang
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Jianming Sun
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Zhiwei Liu
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Kun Li
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Ming Shi
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| | - Jian Li
- Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao, 066004, China
| |
Collapse
|
13
|
Huang Y, Du Z, Lai Z, Wen D, Huang L, He M, Wu Z, Li H, OuYang H, Wu W, Kan A, Shi M. Single-Nucleus and Spatial Transcriptome Profiling Delineates the Multicellular Ecosystem in Hepatocellular Carcinoma After Hepatic Arterial Infusion Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405749. [PMID: 39686623 PMCID: PMC11791974 DOI: 10.1002/advs.202405749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/08/2024] [Indexed: 12/18/2024]
Abstract
Hepatic arterial infusion chemotherapy (HAIC) has emerged as a promising treatment strategy for hepatocellular carcinoma (HCC), but a detailed understanding of the multicellular ecosystem after HAIC treatment is lacking. Here, we collected tumor samples from treatment-naïve primary and post-HAIC HCC, and integrated single-nucleus RNA sequencing with spatial transcriptomics to characterize the tumor ecosystem in the post-HAIC HCC. Increased fractions and enhanced cellular communication of CD4+ T, CD20+ B, and dendritic cell subtypes were identified in post-HAIC tumors. Moreover, it is substantiated that HAIC promoted tertiary lymphoid structures (TLS) formation, and addressed the roles of TLSs as spatial niches of cellular communication. Specifically, intermediate exhausted CD8+ T cells expressing Granzyme-K and PD-1 (PD-1+CD8+ Tex-int) expanded following HAIC and exhibited a functionally antitumor phenotype. PD-1+CD8+ Tex-int accumulated in the TLS vicinity and disseminated throughout the tumor microenvironment, demonstrating potential as an effective biomarker for HAIC-based treatment in HCC. This study provides valuable resources and biological insights in the cellular underpinnings of HAIC treatment.
Collapse
Affiliation(s)
- YeXing Huang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZeFeng Du
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZhiCheng Lai
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - DongSheng Wen
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - LiChang Huang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - MinKe He
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - ZiChao Wu
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - HuiFang Li
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - HanYue OuYang
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - WenChao Wu
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - Anna Kan
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| | - Ming Shi
- Department of Hepatobiliary OncologySun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerState Key Laboratory of Oncology in South ChinaGuangzhou510060P. R. China
| |
Collapse
|
14
|
Abushanab AK, Mustafa MT, Mousa MT, Albanawi RF, Alkhalaileh RM, Alqudah GN, Abu Zaina RF, Abu Sitta ZA, Almasri IM, Abuquteish D. Immune checkpoint inhibitors plus taxane-based chemotherapy for patients with advanced/metastatic NSCLC: a systematic review and meta-analysis across different PD-L1 expression levels. Expert Rev Anticancer Ther 2025; 25:167-179. [PMID: 39874440 DOI: 10.1080/14737140.2025.2460537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are currently the primary approach for managing NSCLC. However, numerous combination therapies are currently under investigation. Our goal is to investigate the overall efficacy and safety of ICIs and taxane-based chemotherapy. METHODS We conducted a systematic review and meta-analysis, searching web databases for relevant literature. We limited our eligibility to phase II/III randomized clinical trials involving advanced/metastatic NSCLC patients. RESULTS We performed a meta-analysis encompassing nineteen studies derived from sixteen RCTs. For patients with sq-NSCLC PD-L1 ≥ 50%, using ICIs plus taxane significantly improve PFS and OS with HR of 0.58 (95% CI, 0.45-0.74, p < 0.0001) and 0.41 (95% CI, 0.33-0.50, p < 0.00001), respectively. For patients with non-sq NSCLC PD-L1 1-49%, the analysis revealed significant improvement of OS and PFS with HR of 0.64 (95% CI, 0.47-0.88, p = 0.005) and 0.62 (95% CI, 0.47-0.81, p = 0.0004), respectively. For TRAEs of all grades, ICIs plus taxane resulted with no significant difference compared to control group with risk ratio (RR) 1.00 (95% CI 0.99-1.02). CONCLUSION The analysis revealed significant improvement in efficacy of ICIs with taxane in advanced/metastatic NSCLC patients compared with ICI/taxane monotherapy.Registration: PROSPERO (CRD42023447532).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dua Abuquteish
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
15
|
Saeheng T, Karbwang J, Na-bangchang K. Immunomodulatory Effects of Atractylodes lancea in Healthy Volunteers with Dosage Prediction for Cholangiocarcinoma Therapy: A Modelling Approach. Pharmaceuticals (Basel) 2025; 18:198. [PMID: 40006012 PMCID: PMC11860138 DOI: 10.3390/ph18020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background and Aims: According to a recent study on the immunomodulatory activity of Atractylodes lancea (Thunb.) DC. (AL) in healthy Thai subjects, AL significantly inhibited the production of key pro-inflammatory cytokines while stimulating the production of immune cells. However, no maximum tolerated dose (MTD) and phase 2A dosage regimens were reported. The study aimed to evaluate the immunomodulatory effects of Atractylodes lancea (Thunb.) DC. (AL) in healthy subjects, and to recommend optimal dose regimens for intrahepatic cholangiocarcinoma (iCCA) based on toxicity criteria. Methods: A physiologically based pharmacokinetic (PBPK) model, combined with the toxicological approach and the immunomodulatory effect, was used for dose-finding. The safety and efficacy of each AL regimen were evaluated based on the previous study. At least a once-daily dose of 1000 mg AL significantly suppressed the production of all pro-inflammatory cytokines while significantly increasing the number of peripheral immune cells. Results: The developed PBPK model predicted the clinically observed data well. No significant differences in SII index values were found, but a difference in the lymphocyte-monocyte ratio was found on day 4. The dosage regimen for phase 2A is a once-daily dose of 1500 or 2000 mg. Preliminary results in phase 2A revealed that a once-daily dose of 2000 mg had a significantly higher median overall survival, progression-free survival, disease control rate, and inhibition of increased tumor size without toxicities compared with control. Conclusions: A PBPK model, in conjunction with a toxicological approach, could assist in finding the potential dosage regimens for a clinical study, including herbal medicine.
Collapse
Affiliation(s)
- Teerachat Saeheng
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
- Graduate Program in Bioclinical Science, Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
| | - Juntra Karbwang
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
| | - Kesara Na-bangchang
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
- Graduate Program in Bioclinical Science, Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Rangsit Campus, Pathumthani 12120, Thailand
| |
Collapse
|
16
|
Li J, Zhang Y, Hu L, Ye H, Yan X, Li X, Li Y, Ye S, Wu B, Li Z. T-cell Receptor Repertoire Analysis in the Context of Transarterial Chemoembolization Synergy with Systemic Therapy for Hepatocellular Carcinoma. J Clin Transl Hepatol 2025; 13:69-83. [PMID: 39801788 PMCID: PMC11712086 DOI: 10.14218/jcth.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/16/2025] Open
Abstract
T-cell receptor (TCR) sequencing provides a novel platform for insight into and characterization of intricate T-cell profiles, advancing the understanding of tumor immune heterogeneity. Recently, transarterial chemoembolization (TACE) combined with systemic therapy has become the recommended regimen for advanced hepatocellular carcinoma. The regulation of the immune microenvironment after TACE and its impact on tumor progression and recurrence has been a focus of research. By examining and tracking fluctuations in the TCR repertoire following combination treatment, novel perspectives on the modulation of the tumor microenvironment post-TACE and the underlying mechanisms governing tumor progression and recurrence can be gained. Clarifying the distinctive metrics and dynamic alterations of the TCR repertoire within the context of combination therapy is imperative for understanding the mechanisms of anti-tumor immunity, assessing efficacy, exploiting novel treatments, and further advancing precision oncology in the treatment of hepatocellular carcinoma. In this review, we initially summarized the fundamental characteristics of TCR repertoire and depicted immune microenvironment remodeling after TACE. Ultimately, we illustrated the prospective applications of TCR repertoires in TACE combined with systemic therapy.
Collapse
Affiliation(s)
- Jie Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Luqi Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Heqing Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xingli Yan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Xin Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Yifan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Shuwen Ye
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Bailu Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Engineering Technology Research Center for Minimally Invasive Interventional Tumors of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
17
|
Miranda I, Jahan N, Shevde LA. The metastatic cascade through the lens of therapeutic inhibition. Cell Rep Med 2025; 6:101872. [PMID: 39706193 DOI: 10.1016/j.xcrm.2024.101872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/23/2024]
Abstract
Metastasis is a main cause of cancer-related death, and a deeper understanding of the metastatic process will inform more targeted and mechanistic approaches that can abrogate challenges in treatment efficacy and toxicity. Several steps throughout the metastatic cascade, from angiogenesis to secondary tumor formation, offer specific vulnerabilities to therapies that can lead to the decline or cessation of metastatic progression. A deeper understanding of the metastatic cascade also allows combination systemic therapies to be used synergistically. In this review, we describe current treatment modalities in the context of multiple steps of the metastatic cascade. We highlight their mechanisms and present their efficacy across multiple cancers. This work also presents targets within the metastatic cascade in need of more research that can advance the landscape of treatments and lead to the goal of metastatic cancer remission.
Collapse
Affiliation(s)
- Ian Miranda
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
18
|
Mo S, Zhong H, Dai W, Li Y, Qi B, Li T, Cai Y. ERBB3-related gene PBX1 is associated with prognosis in patients with HER2-positive breast cancer. BMC Genom Data 2025; 26:2. [PMID: 39794693 PMCID: PMC11720925 DOI: 10.1186/s12863-024-01292-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND HER2-positive breast cancer (BC) is a subtype of breast cancer. Increased ERBB3 expression has been implicated as a potential cause of resistance to other HER-targeted therapies. Our study aimed to screen and validate prognostic markers associated with ERBB3 expression by bioinformatics and affecting the prognosis of HER2 staging. METHODS Analyzing differences in ERBB3-related groups. ERBB3 expression-related differentially expressed genes (DEGs) were identified and intersected with survival status-related DEGs to obtain intersected genes. Three algorithms, LASSO, RandomForest and XGBoost were combined to identify the signature genes. we construct risk models and generate ROC curves for prediction. Furthermore, we delve into the immunological traits, correlations, and expression patterns of signature genes by conducting a comprehensive analysis that encompasses immune infiltration analysis, correlation analysis, and differential expression analysis. RESULTS Significant variability in ERBB3 expression and prognosis in high and low ERBB3 expression groups. Twenty-five candidate DEGs were identified by intersecting ERBB3-related DEGs with survival-related DEGs. Utilizing three distinct machine learning algorithms, we identified three signature genes-PBX1, IGHM, and CXCL13-that exhibited significant diagnostic value within the diagnostic model. In addition, the risk model had better prognostic and predictive effects, and the immune infiltration analysis showed that IGHM, CXCL13 might affect the proliferation of BC cells through immune cells. Functional studies demonstrated that interference with PBX1 inhibited the proliferation, migration, and epithelial-mesenchymal transition process of HER2-positive BC cells. CONCLUSION PBX1, IGHM and CXCL13 are associated with the expression level of the ERBB3 and are prognostic markers for HER2-positive in BC, which may play an important role in the development and progression of BC.
Collapse
Affiliation(s)
- Shufen Mo
- Medical Oncology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China
| | - Haiming Zhong
- Medical Oncology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China
| | - Weiping Dai
- Pathology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China
| | - Yuanyuan Li
- Medical Oncology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China
| | - Bin Qi
- Pathology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China
| | - Taidong Li
- Thoracic Surgery, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China.
- Central Hospital of Guangdong Provincial Nongken, No.2 Renmin Avenue Middle, Xiashan District, Zhanjiang, Guangdong, 524002, China.
| | - Yongguang Cai
- Medical Oncology, Central Hospital of Guangdong Provincial Nongken, Zhanjiang, Guangdong, China.
- Central Hospital of Guangdong Provincial Nongken, No.2 Renmin Avenue Middle, Xiashan District, Zhanjiang, Guangdong, 524002, China.
| |
Collapse
|
19
|
Giuliani J, Tebano U, Mandarà M, Franceschetto A, Giorgi C, Missiroli S, Gabbani M, Napoli G, Luca N, Mangiola D, Muraro M, Perrone M, Pinton P, Fiorica F. "Add More Arrows to Your Quiver": The Role of Adding Another Chemotherapy Drug to Fluoropyrimidine and Long Term Radiotherapy in Locally Advanced Rectal Cancer: A Systematic Review and Meta-Analysis. J Clin Med 2025; 14:345. [PMID: 39860350 PMCID: PMC11765640 DOI: 10.3390/jcm14020345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Objectives: Despite optimal local control obtained with neoadjuvant chemoradiotherapy (CRT), data on overall survival (OS) and disease-free survival (DFS) of local advanced rectal cancer patients are still equivocal. This meta-analysis aimed to estimate the pathological complete response (pCR), regression rate, DFS, and OS probabilities of rectal cancer patients treated with a second chemotherapy drug added to fluoropyrimidine and long-term radiotherapy. Methods: Computerized bibliographic searches of MEDLINE, PUBMED, Web of Science and the Cochrane Central Register of Controlled Trials databases (1970-2023) were supplemented with hand searches of reference lists. Studies were included if they were randomised controlled trials (RCTs) comparing intensified chemotherapy with CRT to preoperative CRT and if they had patients with resectable, histologically proven rectal adenocarcinoma without metastases. Results: Eighteen RCTs (7695 patients) were analysed. Data on population, intervention, and outcomes were extracted from each RCT, following the intention-to-treat method, by three independent observers and combined using the DerSimonian and Laird methods. A chemotherapy with two drug and long-term radiotherapy CRT, compared to preoperative CRT (fluoropyrimidine and long-term radiotherapy), significantly increases the rate of pathological complete response (OR 1.37 (95% CI, 1.16-1.63) p = 0.0003) and the regression rate (OR 1.57 (95% CI, 1.16-2.14) p < 0.00001). Furthermore, it increases DFS (HR 0.87 (95% CI, 0.79 to 0.95) p = 0.002 and OS HR 0.84 (95% CI, 0.74 to 0.95) p = 0.007). The risk of severe adverse events (≥G3) is increased OR 1.96 (95% CI 1.35-2.85), p = 0.0005. Conclusions: In patients with resectable rectal cancer, intensified chemotherapy can reduce by 13% the risk of disease progression and by 16% the risk of death.
Collapse
Affiliation(s)
- Jacopo Giuliani
- Department of Clinical Oncology, Section of Medical Oncology, AULSS 9 Scaligera, 37045 Legnago, Italy; (J.G.); (M.M.); (D.M.)
| | - Umberto Tebano
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Marta Mandarà
- Department of Clinical Oncology, Section of Medical Oncology, AULSS 9 Scaligera, 37045 Legnago, Italy; (J.G.); (M.M.); (D.M.)
| | - Antonella Franceschetto
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (S.M.); (M.P.); (P.P.)
| | - Sonia Missiroli
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (S.M.); (M.P.); (P.P.)
| | - Milena Gabbani
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Giuseppe Napoli
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Nicoletta Luca
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Daniela Mangiola
- Department of Clinical Oncology, Section of Medical Oncology, AULSS 9 Scaligera, 37045 Legnago, Italy; (J.G.); (M.M.); (D.M.)
| | - Marco Muraro
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| | - Mariasole Perrone
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (S.M.); (M.P.); (P.P.)
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 48033 Ferrara, Italy; (C.G.); (S.M.); (M.P.); (P.P.)
| | - Francesco Fiorica
- Department of Clinical Oncology, Section of Medical Oncology, AULSS 9 Scaligera, 37045 Legnago, Italy; (J.G.); (M.M.); (D.M.)
- Department of Clinical Oncology, Section of Radiation Oncology and Nuclear Medicine, AULSS 9 Scaligera, 37045 Legnago, Italy; (U.T.); (A.F.); (M.G.); (G.N.); (N.L.); (M.M.)
| |
Collapse
|
20
|
Hoffmann S, Hoffmann T, Potkrajcic V, Deinzer CKW, Benzler K, Zender L, Daigeler A, Thiel JT. SarkoLife: quality of life in patients undergoing multimodal soft tissue sarcoma treatment. World J Surg Oncol 2025; 23:10. [PMID: 39773479 PMCID: PMC11708100 DOI: 10.1186/s12957-024-03632-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To assess the tolerability of multimodal therapy in soft tissue sarcoma patients, particularly with regard to their quality of life and level of distress. MATERIALS AND METHODS A retrospective cohort study enrolled individuals receiving sarcoma therapy at the sarcoma center of the University of Tuebingen between 2017 and 2022. Participants completed an online survey that included the EORTC's questionnaire (QLQ-C30), coupled with the distress thermometer and demographic inquiries. The primary emphasis was on comparing three distinct modalities: Radiation, Chemotherapy and Surgery. The data were analysed performing one-way ANOVA. RESULTS A total of 237 patients were included in the study. There was a significant difference (p < 0.001) in quality of life according to the EORTC scores (high score = high quality of life) between the different treatments: chemotherapy (mean: 26.8 [standard deviation: 19.5]), radiotherapy (51.0 [21.5]), and surgery (46.9 [28.3]). Similarly, a statistically significant discrepancy (p < 0.001) was found in average distress levels (high score = high level of distress) corresponding to each treatment type: radiation (5.0 [2.7]), surgery (6.0 [2.9]), and chemotherapy (7.4 [2.4]). The rates of patients willing to undergo the same treatment varied across groups, with the highest percentage observed in the surgery group (94.2%), followed by radiation (87.4%), and chemotherapy (73.5%). CONCLUSION Patients receiving multimodal therapy for soft tissue often find chemotherapy particularly demanding. Impairment of both quality of life and physical well-being is more likely and tends to be more severe compared with radiation or surgery. These observations should be taken into consideration when consenting patients and offering treatment plans.
Collapse
Affiliation(s)
- Sebastian Hoffmann
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, Tuebingen, 72076, Germany
| | - Tabea Hoffmann
- Department of Marketing, Faculty of Economics and Business, University Groningen, Groningen, The Netherlands
| | - Vlatko Potkrajcic
- Department of Radiation Oncology, University Hospital Tübingen, Hoppe-Seyler-Str. 3, Tuebingen, 72076, Germany
| | - Christoph K W Deinzer
- Department of Medical Oncology and Pneumology, University Hospital Tuebingen, Otfried-Mueller-Straße 10, Tuebingen, 72076, Germany
| | - Katrin Benzler
- Department of Medical Oncology and Pneumology, University Hospital Tuebingen, Otfried-Mueller-Straße 10, Tuebingen, 72076, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, University Hospital Tuebingen, Otfried-Mueller-Straße 10, Tuebingen, 72076, Germany
- University of Tübingen iFIT Cluster of Excellence (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adrien Daigeler
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, Tuebingen, 72076, Germany
| | - Johannes Tobias Thiel
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, University of Tuebingen, Schnarrenbergstraße 95, Tuebingen, 72076, Germany.
| |
Collapse
|
21
|
Stinson JA, Sheen A, Lax BM, Yang GN, Duhamel L, Santollani L, Fink E, Palmeri J, Wittrup KD. Tumor Integrin-Targeted Glucose Oxidase Enzyme Promotes ROS-Mediated Cell Death that Combines with Interferon Alpha Therapy for Tumor Control. Mol Cancer Ther 2025; 24:118-130. [PMID: 39382078 PMCID: PMC11695183 DOI: 10.1158/1535-7163.mct-24-0163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/14/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Although heightened intratumoral levels of reactive oxygen species (ROS) are typically associated with a suppressive tumor microenvironment, under certain conditions ROS contribute to tumor elimination. Treatment approaches, including some chemotherapy and radiation protocols, increase cancer cell ROS levels that influence their mechanism of cell death and subsequent recognition by the immune system. Furthermore, activated myeloid cells rapidly generate ROS upon encounter with pathogens or infected cells to eliminate disease, and recently, this effector function has been noted in cancer contexts as well. Collectively, ROS-induced cancer cell death may help initiate adaptive antitumor immune responses that could synergize with current approved immunotherapies, for improved control of solid tumors. In this work, we explore the use of glucose oxidase, an enzyme which produces hydrogen peroxide, a type of ROS, to therapeutically mimic the endogenous oxidative burst from myeloid cells to promote antigen generation within the tumor microenvironment. We engineer the enzyme to target pan-tumor-expressed integrins both as a tumor-agnostic therapeutic approach and as a strategy to prolong local enzyme activity following intratumoral administration. We found the targeted enzyme potently induced cancer cell death and enhanced cross-presentation by dendritic cells in vitro and further combined with interferon alpha for long-term tumor control in murine MC38 tumors in vivo. Optimizing the single-dose administration of this enzyme overcomes limitations with immunogenicity noted for other prooxidant enzyme approaches. Overall, our results suggest ROS-induced cell death can be harnessed for tumor control and highlight the potential use of designed enzyme therapies alongside immunotherapy against cancer.
Collapse
Affiliation(s)
- Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brianna M. Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Grace N. Yang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Elizabeth Fink
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Joseph Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
22
|
Jiménez-Cortegana C, Sánchez-Jiménez F, De La Cruz-Merino L, Sánchez-Margalet V. Role of Sam68 in different types of cancer (Review). Int J Mol Med 2025; 55:3. [PMID: 39450529 PMCID: PMC11537268 DOI: 10.3892/ijmm.2024.5444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Src‑associated in mitosis 68 kDa protein (Sam68) is a protein encoded by the heteronuclear ribonucleoprotein particle K homology (KH) single domain‑containing, RNA‑binding, signal transduction‑associated protein 1 (known as KHDRBS1) gene in humans. This protein contains binding sites for critical components in a variety of cellular processes, including the regulation of gene expression, RNA processing and cell signaling. Thus, Sam68 may play a role in a variety of diseases, including cancer. Sam68 has been widely demonstrated to participate in tumor cell proliferation, progression and metastasis to be involved in the regulation of cancer stem cell self‑renewal. Based on the body of evidence available, Sam68 emerges as a promising target for this disease. The objectives of the present included summarizing the role of Sam68 in cancer murine models and cancer patients, unraveling the molecular mechanisms underlying its oncogenic potential and discussing the effectiveness of antitumor agents in reducing the malignant effects of Sam68 during tumorigenesis.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
- Medical Oncology Service, Virgen Macarena University Hospital, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, Consejo Superior de Investigaciones Científicas, University of Seville, 41013 Seville, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, Consejo Superior de Investigaciones Científicas, University of Seville, 41013 Seville, Spain
| |
Collapse
|
23
|
Rinnerthaler G, Egle D, Bartsch R, Schmitt CA, Petzer A, Balic M, Petru E, Denison U, Singer CF, Bjelic-Radisic V, Gampenrieder SP, Knauer M, Sotlar K, Brunner C, Posch F, Hlauschek D, Sölkner L, Bago-Horvath Z, Filipits M, Gili M, Ritter M, Wieser V, Albertini C, Zaborsky N, Weiss L, Marhold M, Schneeweiss B, Pusch R, Gnant M, Greil R. Neoadjuvant atezolizumab in combination with dual HER2 blockade plus epirubicin in women with early HER2-positive breast cancer: the randomized phase 2 ABCSG-52/ATHENE trial. NATURE CANCER 2025; 6:41-50. [PMID: 39820125 PMCID: PMC11779624 DOI: 10.1038/s43018-024-00890-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025]
Abstract
The role of anthracyclines in the treatment of early breast cancer (EBC) is increasingly being challenged, especially in de-escalation strategies. However, owing to their immunogenic effects, anthracyclines are promising combination partners with immunotherapies. In the randomized phase 2 trial ABCSG-52 (EudraCT no. 2019-002364-27), we investigated epirubicin plus immunotherapy in women with human epidermal growth factor receptor 2 (HER2)-positive EBC. A total of 58 patients were randomized 1:1 to two cycles of a chemotherapy-free induction phase (part 1) of dual HER2 blockade with trastuzumab and pertuzumab (TP) plus the anti-programmed death ligand 1 antibody atezolizumab (TP-A) or TP alone. Thereafter, all patients received four cycles of TP-A in combination with epirubicin (part 2). The primary endpoint, pathological complete response (pCR), was met in 35 patients (60.3%; 95% confidence interval (CI) 47.5% to 71.9%), 19 patients (65.5%) in the TP-A group and 16 patients (55.2%) in the TP group. The residual cancer burden 0/I rate and objective response rate (secondary endpoints) in all patients with evaluable data were 80.0% (n = 44/55; 95% CI 67.6% to 88.4%) and 89.3% (n = 50/56; 95% CI 78.5% to 95.0%), respectively. Grade ≥3 adverse events were reported in 17 patients (29.3%). Based on our findings, we conclude that a neoadjuvant chemotherapy de-escalation immunotherapy regimen with trastuzumab, pertuzumab, atezolizumab and epirubicin is effective and safe in patients with HER2-positive EBC.
Collapse
Affiliation(s)
- Gabriel Rinnerthaler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, Austria.
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria.
| | - Daniel Egle
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Rupert Bartsch
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Division of Oncology, Department of Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Clemens A Schmitt
- Department of Hematology and Internal Oncology, Johannes Kepler University, Kepler University Hospital, Linz, Austria
| | - Andreas Petzer
- Internal Medicine I: Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Austria
| | - Marija Balic
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Edgar Petru
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Ursula Denison
- Institute for Gynecological Oncology and Senology, Karl Landsteiner Society, Hietzing Hospital, Vienna, Austria
| | - Christian F Singer
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Vesna Bjelic-Radisic
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Breast Unit, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - Simon Peter Gampenrieder
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, Austria
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Michael Knauer
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Tumor and Breast Center Eastern Switzerland, St. Gallen, Switzerland
| | - Karl Sotlar
- Department of Pathology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Christine Brunner
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dominik Hlauschek
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Lidija Sölkner
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Martin Filipits
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Manuela Gili
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Magdalena Ritter
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Wieser
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Carmen Albertini
- Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadja Zaborsky
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, Austria
- Laboratory of Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria
| | - Lukas Weiss
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, Austria
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Maximilian Marhold
- Division of Oncology, Department of Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Bruno Schneeweiss
- Department of Hematology and Internal Oncology, Johannes Kepler University, Kepler University Hospital, Linz, Austria
| | - Renate Pusch
- Internal Medicine I: Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Austria
| | - Michael Gnant
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Richard Greil
- Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, Austria
- Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| |
Collapse
|
24
|
Li Y, Li B, Chen C, Hou H, Su M, Li F, Xiao Z, Yang X. Capturing Hydrophilic Chemotherapeutics Agents Into siRNA-Encapsulated Vesicle-Like Nanoparticles for Convenient ICB-Chemo Combination Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404073. [PMID: 39498748 DOI: 10.1002/smll.202404073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/15/2024] [Indexed: 11/07/2024]
Abstract
Clinical evidence has demonstrated that combining immune checkpoint blockade (ICB) therapy with chemotherapy significantly improves response rates to ICB therapy and therapeutic efficacy in various tumor types. However, a convenient method for achieving synergistic ICB therapy and chemotherapy with precise co-delivery of both agents is still highly desirable. In this study, a strategy for co-delivering small interfering RNA (siRNA) encapsulated in vesicle-like nanoparticles (VNPsiRNA) and chemotherapeutic drugs is aimed to develop. It is discovered that the hydrophilic chemotherapeutic drug mitoxantrone hydrochloride (MTO·2HCl) can be captured into VNPsiRNA. The resulting VNPsiRNACpMTO can simultaneously block immune checkpoints via RNA silencing and induce chemotherapeutic effects on tumor cells. The mechanism of MTO·2HCl is elucidates, captures, and demonstrates the superior therapeutic effect of VNPsiRNACpMTO through chemo-immunotherapy. This strategy can also be extended to deliver other hydrochloride anticancer drugs, such as doxorubicin hydrochloride (DOX·HCl), for achieving synergistic combination therapy. This study provides a facile strategy for enhancing combined ICB and chemotherapy via co-delivering siRNA and chemotherapeutic drugs, offering a promising approach to cancer treatment.
Collapse
Affiliation(s)
- Yan Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Bingqin Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Chaoran Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Hengliang Hou
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Miao Su
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Fangzheng Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Zekai Xiao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| |
Collapse
|
25
|
Koganemaru S, Koyama S, Suto F, Koga M, Inaki K, Kuwahara Y, Arita T, Hirata T, Goto H, Wada N, Kobayashi M, Shibutani T, Okabayashi T, Nakamaru K, Kawazoe A, Togashi Y, Nishikawa H, Shitara K. The Tumor Immune Microenvironment and Therapeutic Efficacy of Trastuzumab Deruxtecan in Gastric Cancer. CANCER RESEARCH COMMUNICATIONS 2025; 5:84-93. [PMID: 39679910 PMCID: PMC11729160 DOI: 10.1158/2767-9764.crc-24-0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/01/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
SIGNIFICANCE This biomarker study explored HER2 expression levels and immune cell characteristics that may affect response to T-DXd using tumor tissue samples collected from clinical trial participants. The results suggest that HER2 expression levels and tumor characteristics before the initiation of T-DXd may correlate with the efficacy of the drug.
Collapse
Affiliation(s)
- Shigehiro Koganemaru
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Chiba, Japan
- Department of Immunogenomic Medicine, Research Institute, National Cancer Center, Tokyo, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumitaka Suto
- Translational Science Department, Precision Medicine Function, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Makito Koga
- Translational Science Department I, Precision Medicine Function, Daiichi Sankyo, Co., Ltd., Tokyo, Japan
| | - Koichiro Inaki
- Oncology Medical Science Department, Medical Affairs Division, Daiichi Sankyo, Co., Ltd., Tokyo, Japan
| | - Yusuke Kuwahara
- Translational Science Department, Precision Medicine Function, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Takeo Arita
- Translational Science Department I, Precision Medicine Function, Daiichi Sankyo, Co., Ltd., Tokyo, Japan
| | - Tsuyoshi Hirata
- Translational Science Department, Precision Medicine Function, Daiichi Sankyo, Inc., Basking Ridge, New Jersey
| | - Hiroki Goto
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan
| | - Naoya Wada
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan
| | - Maki Kobayashi
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan
| | - Tomoko Shibutani
- Translational Research Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo, Japan
| | - Tatsuya Okabayashi
- Clinical Development Department I, Development Function, Daiichi Sankyo, Co., Ltd., Tokyo, Japan
| | - Kenji Nakamaru
- Translational Science Department I, Precision Medicine Function, Daiichi Sankyo, Co., Ltd., Tokyo, Japan
| | - Akihito Kawazoe
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yousuke Togashi
- Department of Tumor Microenvironment, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo/Chiba, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohei Shitara
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
26
|
Lefler DS, Manobianco SA, Bashir B. Immunotherapy resistance in solid tumors: mechanisms and potential solutions. Cancer Biol Ther 2024; 25:2315655. [PMID: 38389121 PMCID: PMC10896138 DOI: 10.1080/15384047.2024.2315655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
While the emergence of immunotherapies has fundamentally altered the management of solid tumors, cancers exploit many complex biological mechanisms that result in resistance to these agents. These encompass a broad range of cellular activities - from modification of traditional paradigms of immunity via antigen presentation and immunoregulation to metabolic modifications and manipulation of the tumor microenvironment. Intervening on these intricate processes may provide clinical benefit in patients with solid tumors by overcoming resistance to immunotherapies, which is why it has become an area of tremendous research interest with practice-changing implications. This review details the major ways cancers avoid both natural immunity and immunotherapies through primary (innate) and secondary (acquired) mechanisms of resistance, and it considers available and emerging therapeutic approaches to overcoming immunotherapy resistance.
Collapse
Affiliation(s)
- Daniel S. Lefler
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven A. Manobianco
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Babar Bashir
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
27
|
Ogasawara M. Wilms' tumor 1 -targeting cancer vaccine: Recent advancements and future perspectives. Hum Vaccin Immunother 2024; 20:2296735. [PMID: 38148629 PMCID: PMC10760787 DOI: 10.1080/21645515.2023.2296735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023] Open
Abstract
This mini-review explores recent advancements in cancer vaccines that target Wilms' tumor (WT1). Phase I/II trials of WT1 peptide vaccines have demonstrated their safety and efficacy against various cancers. Early trials employing HLA class I peptides evolved through their combination with HLA class II peptides, resulting in improved clinical outcomes. Additionally, WT1-targeted dendritic cell vaccines have exhibited favorable results. Studies focusing on hematological malignancies have revealed promising outcomes, including long-term remission and extended survival times. The combination of vaccines with immune checkpoint inhibitors has shown synergistic effects. Current preclinical developments are focused on enhancing the effectiveness of WT1 vaccines, underscoring the necessity for future large-scale Phase III trials to further elucidate their efficacy.
Collapse
Affiliation(s)
- Masahiro Ogasawara
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan
| |
Collapse
|
28
|
Zanchetta C, De Marchi L, Macerelli M, Pelizzari G, Costa J, Aprile G, Cortiula F. Antibody-Drug Conjugates in Non-Small Cell Lung Cancer: State of the Art and Future Perspectives. Int J Mol Sci 2024; 26:221. [PMID: 39796075 PMCID: PMC11719753 DOI: 10.3390/ijms26010221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent one of the most promising and rapidly emerging anti-cancer therapies because they combine the cytotoxic effect of the conjugate payload and the high selectivity of the monoclonal antibody, which binds a specific membrane antigen expressed by the tumor cells. In non-small cell lung cancer (NSCLC), ADCs are being investigated targeting human epidermal growth factor receptor 2 (HER2), human epidermal growth factor receptor 3 (HER3), trophoblast cell surface antigen 2 (TROP2), Mesenchymal-epithelial transition factor (c-MET), and carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5). To date, Trastuzumab deruxtecan is the only ADC that has been approved by the FDA for the treatment of patients with NSCLC, but several ongoing studies, both using ADCs as monotherapy and combined with other therapies, are investigating the efficacy of new ADCs. In this review, we describe the structures and mechanism of action of different ADCs; we present the evidence derived from the main clinical trials investigating ADCs' efficacy, focusing also on related toxicity; and, finally, we discuss future perspectives in terms of toxicity management, possible biomarkers, and the identification of resistance mechanisms.
Collapse
Affiliation(s)
- Carol Zanchetta
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (C.Z.); (L.D.M.); (J.C.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Lorenzo De Marchi
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (C.Z.); (L.D.M.); (J.C.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Marianna Macerelli
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Giacomo Pelizzari
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Jacopo Costa
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (C.Z.); (L.D.M.); (J.C.)
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Giuseppe Aprile
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
| | - Francesco Cortiula
- Department of Oncology, University Hospital of Udine, 33100 Udine, Italy; (M.M.); (G.P.); (G.A.)
- Department of Respiratory Medicine, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
29
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2770-x. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
30
|
Kuo YC, Tai TS, Yang HY, Lui KW, Chao YK, Lee LY, Huang Y, Fan HC, Lin AC, Hsieh CH, Yang Z, Chang KP, Lin CY, Wang HM, Hsu CL. Characterization of the immune cell profile in metastatic nasopharyngeal carcinoma treated with chemotherapy and immune checkpoint inhibitors. Am J Cancer Res 2024; 14:5717-5733. [PMID: 39803661 PMCID: PMC11711527 DOI: 10.62347/sspi9013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated cancer, and immune checkpoint inhibitors (ICIs) have shown efficacy in its treatment. The combination of chemotherapy and ICIs represents a new trend in the standard care for metastatic NPC. In this study, we aim to clarify the immune cell profile and related prognostic factors in the ICI-based treatment of metastatic NPC. Programmed cell death ligand 1 (PD-L1) expression was measured in 81 metastatic tissue samples that had not received prior ICI treatment. The combined positive score (CPS) was positive in 58.0% of the samples, with a statistically significant correlation to median overall survival (OS) (CPS ≥ 1 vs. CPS < 1: 28 vs. 16 months, P = 0.004). For the combination treatment of metastatic NPC, 62 patients were enrolled in a retrospective analysis, yielding a median OS of 39.3 months. The objective response rate for this combination therapy was 71%, with a complete response rate of 45.2%. With a cutoff value of 4.8 for the pre-treated neutrophil-lymphocyte ratio (NLR) in peripheral blood (PB), the difference in median OS was statistically significant (P = 0.021). Thirty-seven patients received local treatment following the combination therapy of ICIs and chemotherapy, which provided additional survival benefits. Most hyper-responders exhibited a prolonged low NLR (< 3), a high total lymphocyte count, and an undetectable or stable EBV DNA load in PB during treatment. Peripheral blood mononuclear cells (PBMCs) from most patients receiving the combination treatment were rich in PD-1+CD8+ lymphocytes, which showed high expression of both IFN-γ and Granzyme B, demonstrating the ability to kill the EBV-positive NPC cell line and xenografts in vitro and in vivo. Responders also displayed increased levels of CD4+CD45RA-CCR7-CD28+CD57- cells (effector memory cell subset) in peripheral blood. These results indicate that in the context of combined chemotherapy and ICIs, high PD-L1 expression in pre-treated metastatic tumor tissue, a low NLR before treatment, a decrease in NLR after treatment, and local treatment can provide significant benefits for patients with metastatic NPC.
Collapse
Affiliation(s)
- Yung-Chia Kuo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Tzong-Shyuan Tai
- Advanced Immunology Laboratory, Chang Gung Memorial HospitalLinkou, Taoyuan 33305, Taiwan
| | - Huang-Yu Yang
- Advanced Immunology Laboratory, Chang Gung Memorial HospitalLinkou, Taoyuan 33305, Taiwan
- Kidney Research Institute, Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Kar-Wai Lui
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Yin-Kai Chao
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
- School of Medicine, National Tsing-Hua UniversityHsinchu 300044, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial HospitalLinkou, Taoyuan 33305, Taiwan
| | - Hsien-Chi Fan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - An-Chi Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Zhangung Yang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Chien-Yu Lin
- Department of Radiation, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Hung-Ming Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
| | - Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung UniversityTaoyuan 33305, Taiwan
- School of Medicine, Chang Gung UniversityTaoyuan 33305, Taiwan
| |
Collapse
|
31
|
Koshrovski-Michael S, Ajamil DR, Dey P, Kleiner R, Tevet S, Epshtein Y, Green Buzhor M, Khoury R, Pozzi S, Shenbach-Koltin G, Yeini E, Woythe L, Blau R, Scomparin A, Barshack I, Florindo HF, Lazar S, Albertazzi L, Amir RJ, Satchi-Fainaro R. Two-in-one nanoparticle platform induces a strong therapeutic effect of targeted therapies in P-selectin-expressing cancers. SCIENCE ADVANCES 2024; 10:eadr4762. [PMID: 39671487 PMCID: PMC11641104 DOI: 10.1126/sciadv.adr4762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Combined therapies in cancer treatment aim to enhance antitumor activity. However, delivering multiple small molecules imposes challenges, as different drugs have distinct pharmacokinetic profiles and tumor penetration abilities, affecting their therapeutic efficacy. To circumvent this, poly(lactic-co-glycolic acid) (PLGA)-polyethylene glycol (PEG)-based nanoparticles were developed as a platform for the codelivery of synergistic drug ratios, improving therapeutic efficacy by increasing the percentage of injected dose reaching the tumor. Nonetheless, extravasation-dependent tumor accumulation is susceptible to variations in tumor vasculature; therefore, PLGA-PEG was modified with sulfates to actively target P-selectin-expressing cancers. Here, we show the potential of our platform in unique three-dimensional (3D) in vitro and in vivo models. The P-selectin-targeted nanoparticles showed enhanced accumulation in 3D spheroids and tissues of P-selectin-expressing BRAF-mutated melanomas and BRCA-mutated breast cancers, resulting in superior in vivo efficacy and safety. This nanoplatform could advance the codelivery of a plethora of anticancer drug combinations to various P-selectin-expressing tumors.
Collapse
Affiliation(s)
- Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Chemistry, Siksha Bhavana, Visva-Bharati University, Santiniketan, West Bengal 731235, India
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shahar Tevet
- The Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- ADAMA Center for Novel Delivery Systems in Crop Protection, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Green Buzhor
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Laura Woythe
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology (TU/e), 5612AZ Eindhoven, Netherlands
| | - Rachel Blau
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Turin 10125, Italy
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon 1649-003, Portugal
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute of Biological Research (IIBR), Ness-Ziona 74100, Israel
| | - Lorenzo Albertazzi
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology (TU/e), 5612AZ Eindhoven, Netherlands
| | - Roey J. Amir
- The Center for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Organic Chemistry, School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- ADAMA Center for Novel Delivery Systems in Crop Protection, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
32
|
Obeagu EI, Obeagu GU. Lymphocyte infiltration in breast cancer: A promising prognostic indicator. Medicine (Baltimore) 2024; 103:e40845. [PMID: 39654199 PMCID: PMC11631027 DOI: 10.1097/md.0000000000040845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/09/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Breast cancer is a leading cause of cancer-related mortality among women worldwide, necessitating the identification of reliable prognostic markers to guide treatment and improve patient outcomes. Recent research has highlighted the prognostic significance of tumor-infiltrating lymphocytes (TILs) in breast cancer, with high levels of TILs being associated with improved survival rates and better responses to therapy. This review delves into the mechanisms driving lymphocyte infiltration, its clinical implications, and the potential for TILs to serve as predictive biomarkers in breast cancer management. The presence of TILs within the tumor microenvironment reflects a dynamic interplay between tumor cells and the host immune system. Chemokine signaling, antigen presentation, and immune checkpoint interactions are key mechanisms that facilitate the recruitment and activity of lymphocytes at the tumor site. Clinically, the density of TILs varies across breast cancer subtypes, with the most significant prognostic value observed in triple-negative and HER2-positive breast cancers. High TIL levels correlate with improved overall survival and disease-free survival, underscoring their potential as a valuable prognostic indicator. Therapeutically, the role of TILs has opened new avenues in breast cancer treatment, particularly in the realm of immunotherapy. Immune checkpoint inhibitors, adoptive cell therapy, and combination therapies leveraging TILs are being explored to enhance antitumor responses. As research progresses, the integration of TIL assessment into routine clinical practice could revolutionize personalized treatment strategies, ultimately improving prognostic accuracy and patient outcomes in breast cancer care.
Collapse
|
33
|
Kurnit KC, Odunsi K. Harnessing Antitumor Immunity in Ovarian Cancer. Cold Spring Harb Perspect Med 2024; 14:a041336. [PMID: 38621830 PMCID: PMC11610759 DOI: 10.1101/cshperspect.a041336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Despite progress in other tumor types, immunotherapy is not yet part of the standard of care treatment for high-grade serous ovarian cancer patients. Although tumor infiltration by T cells is frequently observed in patients with ovarian cancer, clinical responses to immunotherapy remain low. Mechanisms for immune resistance in ovarian cancer have been explored and may provide insight into future approaches to improve response to immunotherapy agents. In this review, we discuss what is known about the immune landscape in ovarian cancer, review the available data for immunotherapy-based strategies in these patients, and provide possible future directions.
Collapse
Affiliation(s)
- Katherine C Kurnit
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois 60637, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois 60637, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois 60637, USA
| |
Collapse
|
34
|
Laface C, Fina E, Ricci AD, Guven DC, Ambrogio F, De Summa S, Vitale E, Massafra R, Brunetti O, Rizzo A. Immunobiology of biliary tract cancer and recent clinical findings in approved and upcoming immune checkpoint inhibitors. Expert Opin Biol Ther 2024; 24:1363-1374. [PMID: 39545466 DOI: 10.1080/14712598.2024.2431088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Recently, immunotherapy has offered new hope for treating biliary tract cancer (BTC). However, several issues are to be considered, including the lack of validated predictive biomarkers that could help to identify patient groups which are most likely to benefit from such therapeutic approaches. AREAS COVERED In the current article, we will provide an overview of recent results and ongoing and future research directions of immunotherapy in BTC, with a special focus on recently published, practice-changing data, and ongoing active and recruiting clinical trials. EXPERT OPINION At this moment, dozens of clinical trials in phases I to III are evaluating the role of cancer immunotherapy in this setting, with the hope of adding more therapeutic options for BTC patients. Future research must focus on the development of novel agents and combinations, but the validation of biomarkers remains an urgent need. As more research results emerge, novel combinatorial strategies are destined to further transform the treatment paradigm for this heterogeneous and aggressive tumor type.
Collapse
Affiliation(s)
- Carmelo Laface
- Azienda Sanitaria Provinciale, Reggio Calabria (RC), Italy
| | - Emanuela Fina
- Thoracic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
- Medical Oncology Clinic, Elazig City Hospital, Health Sciences University, Elazig, Turkey
| | - Francesca Ambrogio
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori, "Giovanni Paolo II", Bari, Italy
| | - Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
35
|
Elgohary MK, Elkotamy MS, Al-Warhi T, Eldehna WM, Abdel-Aziz HA. Development of new LSM-83177 analogues as anti-tumor agents against colorectal cancer targeting p53-MDM2 interaction. Bioorg Chem 2024; 153:107766. [PMID: 39244969 DOI: 10.1016/j.bioorg.2024.107766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/29/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
LSM-83177, a phenoxy acetic acid derivative, is a small molecule reported for its promising anti-tumor properties. Via inhibiting the interaction between MDM2 and p53, LSM-83177 can elevate the active p53 levels within cells, thereby promoting apoptosis and inhibiting tumor growth. Also, LSM-83177 has been shown to inhibit GST activity in colorectal cancer HT29 cells. In the current work, novel LSM-83177 hydrazone analogs 5a-f, 7a-b, 10a-e, and 13a-b have been designed according to the structure features of LSM-83177 and their binding mode in the active site of MDM2. The anti-cancer activity of the newly synthesized analogs is evaluated against the HT29 cell line. The most potent compounds, 7a and 10a, showed IC50 = 12.48 and 10.44 µg/ml, respectively, when compared with Cisplatin (IC50 = 11.32 µg/ml) as a reference drug. Compounds 7a and 10a were introduced for further inspection for p53-MDM2 protein-protein interaction, where they displayed IC50 values of 3.65 and 11.08 µg/ml, respectively. Furthermore, hydrazones 7a and 10a increased the p-53 expression levels by 3.22- and 4.25-fold, respectively; in addition, they effectively reduced the GST expression levels in HT29 cancer cells with 0.56- and 0.30-fold increments in comparison to the untreated control.
Collapse
Affiliation(s)
- Mohamed K Elgohary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt.
| | - Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo, 11829, Egypt
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt.
| | - Hatem A Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Dokki, Cairo, 12622, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt.
| |
Collapse
|
36
|
Zha C, Yang X, Yang J, Zhang Y, Huang R. Immunosuppressive microenvironment in acute myeloid leukemia: overview, therapeutic targets and corresponding strategies. Ann Hematol 2024; 103:4883-4899. [PMID: 39607487 DOI: 10.1007/s00277-024-06117-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Similar to other malignancies, immune dysregulation is a key feature of acute myeloid leukemia (AML), manifesting as suppressed anti-leukemia immune cells, immune evasion by leukemia blasts, and disease progression. Various immunosuppressive factors within the AML microenvironment contribute to the weakening of host immune responses and the efficacy of cellular immunotherapy. To address these challenges, strategies targeting immunosuppressive elements within the AML microenvironment aim to bolster host or adoptive immune effector cells, ultimately enhancing leukemia treatment. Additionally, the off-target effects of certain targeted drugs (venetoclax, sorafenib, ivosidenib, etc.) may also positively impact anti-AML immunity and immunotherapy. This review provides an overview of the immunosuppressive factors present in AML microenvironment and the strategies developed to rescue immune cells from immunosuppression. We also outline how targeted agents can alter the immune landscape in AML patients, and discuss the potential of targeted drugs to benefit host anti-leukemia immunity and immunotherapy for AML.
Collapse
Affiliation(s)
- Chenyu Zha
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyu Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rui Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
37
|
Wang Z, Jin L, Wang J, Tian X, Mi T, Li M, Zhang Z, Wu X, Li M, Liu J, Wang Z, Liu Y, Luo J, Ren C, He D. Recruitment and polarization typing of tumor-associated macrophages is associated with tumor progression and poor prognosis in Wilms tumor patients. PLoS One 2024; 19:e0309910. [PMID: 39531417 PMCID: PMC11556688 DOI: 10.1371/journal.pone.0309910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/17/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Tumor-associated macrophages (TAMs) play a crucial role in shaping various tumor microenvironments. However, their recruitment in Wilms tumor (WT), the predominant malignant renal tumor in children, has been inadequately explored. This retrospective cohort study involved the analysis of 148 WT samples to investigate the recruitment and polarization typing of TAMs in WT tissues. METHODS WT tissues underwent Western blotting (WB), reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and immunofluorescence (IF) to measure the expression of TAM markers CD68, CD86, and CD163. Statistically analyze the relationship between TAM recruitment levels and patient clinical characteristics, and use Kaplan-Meier curves and the log-rank test to evaluate the association between TAM levels and survival outcomes. RESULTS The findings indicated a positive correlation between the recruitment levels of total macrophages (Mtotal) and M2 tumor-associated macrophages (M2 TAM) in both chemotherapy and non-chemotherapy groups with the clinical stage. Elevated recruitment of Mtotal and M2 TAM in tumor tissues was linked to a poorer prognosis. Notably, patients with persistently higher recruitment of M2 TAM following preoperative chemotherapy exhibited the worst prognosis. CONCLUSIONS The recruitment and polarization typing of TAM exhibit significant differences in WT patients with various stages and prognosis outcomes, suggesting a potential avenue for future diagnosis and treatment of WT.
Collapse
Affiliation(s)
- Zhang Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Liming Jin
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jinkui Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaomao Tian
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tao Mi
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Mujie Li
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhaoxia Zhang
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xin Wu
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Maoxian Li
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jiayan Liu
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhaoying Wang
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yimeng Liu
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Junyi Luo
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chunnian Ren
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Dawei He
- Department of Urology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
38
|
Arai H, Gandhi N, Battaglin F, Wang J, Algaze S, Jayachandran P, Soni S, Zhang W, Yang Y, Millstein J, Lo JH, Sohal D, Goldberg R, Hall MJ, Scott AJ, Hwang JJ, Lou E, Weinberg BA, Marshall J, Goel S, Xiu J, Korn WM, Lenz HJ. Role of CD47 gene expression in colorectal cancer: a comprehensive molecular profiling study. J Immunother Cancer 2024; 12:e010326. [PMID: 39500526 PMCID: PMC11733795 DOI: 10.1136/jitc-2024-010326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/07/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND In patients with colorectal cancer (CRC), the therapeutic effects of conventional immune checkpoint inhibitors targeting the adaptive immune system are largely limited to those with microsatellite instability-high tumors. Meanwhile, new immunotherapies targeting the innate immune system are attracting increasing attention. CD47 is a representative innate immune checkpoint involved in the evasion of tumor cell phagocytosis by macrophages. This large-scale study comprehensively examined the molecular significance of CD47 gene expression in CRC. METHODS We analyzed the next-generation sequencing data of DNA and RNA from 14,287 CRC cases included in the data set of a commercial Clinical Laboratory Improvement Amendments-certified laboratory (Caris Life Sciences). The cases were divided into two groups based on the median value of CD47 gene expression levels. The molecular and immune profiles between the groups were compared, and the relationship between CD47 expression and survival outcomes was further examined. RESULTS In CD47-high tumors, the proportion of consensus molecular subtypes 1 and 4 was significantly higher than in CD47-low tumors. The expression levels of damage-associated molecular pattern-related genes showed a positive correlation with CD47 expression levels. Major oncogenic pathways, such as mitogen-activated protein kinase, phosphoinositide 3-kinase, angiogenesis, and transforming growth factor beta, were significantly activated in CD47-high tumors. Additionally, the expression levels of a panel of adaptive immune checkpoint genes and estimates of immune cells constituting the tumor microenvironment (TME) were significantly higher in CD47-high tumors. CONCLUSIONS CD47 expression in CRC was associated with the activation of several oncogenic pathways and an immune-engaged TME. Our findings may provide valuable information for considering new therapeutic strategies targeting innate immune checkpoints in CRC.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Clinical Oncology, St Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Nishant Gandhi
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Caris Life Sciences, Phoenix, Arizona, USA
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jingyuan Wang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sandra Algaze
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Priya Jayachandran
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yan Yang
- Department of Population and Public Health Sciences, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Davendra Sohal
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Richard Goldberg
- West Virginia University Cancer Institute, Morgantown, West Virginia, USA
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Aaron James Scott
- Department of Medicine, University of Arizona Cancer Center, Tucson, Arizona, USA
| | - Jimmy J Hwang
- Department of Solid Tumor Oncology, GI Medical Oncology Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Benjamin A Weinberg
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - John Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Sanjay Goel
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Joanne Xiu
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Caris Life Sciences, Phoenix, Arizona, USA
| | - W Michael Korn
- Clinical & Translational Research, Medical Affairs, Caris Life Sciences, Caris Life Sciences, Phoenix, Arizona, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
39
|
Xia X, Wang Y, Wang M, Lin J, Wang R, Xie S, Yu Y, Long J, Huang Z, Xian H, Zhang W, Lu C, Wang W, Liu H. The enhancement of immunoactivity induced by immunogenic cell death through serine/threonine kinase 10 inhibition: a potential therapeutic strategy. Front Immunol 2024; 15:1451796. [PMID: 39555062 PMCID: PMC11563836 DOI: 10.3389/fimmu.2024.1451796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/09/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Immunogenic cell death (ICD) is capable of activating the anti-tumor immune response of the organism; however, it is concurrently a complex process involving multiple factors. The specific factors that impact the occurrence of ICD remain undefined. Methods Through cluster analysis, patient specimens retrieved from the TARGET, TCGA, and GEO AML databases were categorized into two subtypes based on the expression levels of ICD-related genes: ICD-high and ICD-low. We compared the prognostic survival outcomes, pathway enrichment analysis, and immune cell infiltration between these two subtypes. Additionally, we identified factors related to AML development from multiple databases and verified the role of these factors both in vivo and in vitro in activating the immune response during the occurrence of ICD. Results and discussion In the ICD-high subtype, there was a notable increase in the abundance of immune cell populations, along with the enrichment of pathways pertinent to the activation of various immune cells. Despite these immunological enhancements, this subgroup demonstrated a poorer prognosis. This phenomenon was consistently observed across various additional AML datasets, leading us to hypothesize that elevated expression of ICD genes does not invariably correlate with a favorable prognosis. Notably, STK10 exhibited elevated expression in AML, was associated with a poor prognosis, and showed synchronous expression patterns with ICD genes. Inhibition of STK10 led to the activation of ICD and the induction of an antitumor response. Moreover, when combined with other ICD inducers, it produced a synergistic anti-tumor effect. Our results reveal the impact of STK10 on ICD and underscore its key role in initiating ICD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Wenfang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital, School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Han Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital, School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
40
|
Lamy D, Mouillot P, Mariet A, Barnestein R, Quilot F, Fraisse C, Ghiringhelli F, Bonniaud P, Zouak A, Foucher P. Real-world comparison of chemo-immunotherapy and chemotherapy alone in the treatment of extensive-stage small-cell lung cancer. Respir Med Res 2024; 86:101125. [PMID: 39033607 DOI: 10.1016/j.resmer.2024.101125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is a high-grade neuroendocrine carcinoma responsible for 200,000 deaths per year worldwide. Platinum-etoposide-based chemotherapy has been the standard of treatment for the past 40 years, with an overall survival of 10 months. Since 2019, the addition of immunotherapy (atezolizumab or durvalumab) to chemotherapy has become the standard of care for first-line treatment of extensive-stage SCLC following the demonstration of an improvement in overall survival in phase 3 studies. We aimed to evaluate the efficacy and safety of chemo-immunotherapy compared with chemotherapy alone in a "real-world" setting. METHODS Retrospective observational study including patients undergoing first-line treatment for extensive-stage SCLC between 2014 and 2022. We separated the study population into two arms (chemo-immunotherapy/chemotherapy). For each arm, progression-free survival (PFS), overall survival (OS) and serious side effects were collected. Associations between treatments and survival outcomes were adjusted for potential confounders. Consolidative palliative thoracic radiotherapy was introduced in the models as a time-dependent variable. RESULTS A total of 118 patients with a median age of 63 years were included. 65.2 % of patients were performance status 0 or 1. In univariate analysis, PFS and OS were not significantly different between the chemo-immunotherapy and chemotherapy alone groups (p = 0.70 and 0.24 respectively). In multivariate analysis, the addition of immunotherapy to chemotherapy was not significantly associated with better PFS (HR 0.76, IC (0.49 - 1.19), p = 0.23), but it was significantly associated with better OS (HR 0.61, IC (0.38 - 0.98), p = 0.04). Consolidative palliative thoracic radiotherapy (time-dependent variable), when applied (almost only in the chemotherapy alone group), was significantly associated with better PFS and OS. DISCUSSION In this real-world study, chemo-immunotherapy was associated with slightly better OS compared to chemotherapy alone as a first-line treatment in ES-SCLC patients in multivariate analysis, which is not explained by a benefit in PFS. However, consolidative palliative thoracic radiotherapy seems to be significantly associated with better OS and PFS, suggesting that we should also consider using it in patients receiving chemo-immunotherapy.
Collapse
Affiliation(s)
- D Lamy
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France
| | - P Mouillot
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France; University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France; INSERM U1231 CTM, Labex LIPSTIC and label of excellence from la Ligue National contre le cancer, France
| | - A Mariet
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France; CHU Dijon Bourgogne, Service de Biostatistiques et d'information médicale, Dijon, France; INSERM, Université de Bourgogne, CHU Dijon Bourgogne, CIC 1432, Module Épidémiologie Clinique, Dijon, France; CHU Dijon-Bourgogne, ResAM, Dijon, France
| | - R Barnestein
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France
| | - F Quilot
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France
| | - C Fraisse
- CHU Dijon-Bourgogne, ResAM, Dijon, France
| | - F Ghiringhelli
- University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France; CHU Dijon-Bourgogne, ResAM, Dijon, France; INSERM U1231 CTM, Labex LIPSTIC and label of excellence from la Ligue National contre le cancer, France
| | - P Bonniaud
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France; University of Burgundy, Faculty of Medicine and Pharmacy, Dijon, France; INSERM U1231 CTM, Labex LIPSTIC and label of excellence from la Ligue National contre le cancer, France.
| | - A Zouak
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France
| | - P Foucher
- Service d'Oncologie Thoracique et Service de Pneumologie et Soins Intensifs Respiratoire, Hôpital Dijon-Bourgogne, Dijon, France
| |
Collapse
|
41
|
Wang K, Leyba A, Hsu R. Addressing the unmet need in NSCLC progression with advances in second-line therapeutics. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1297-1320. [PMID: 39759220 PMCID: PMC11700623 DOI: 10.37349/etat.2024.00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025] Open
Abstract
Lung cancer is the leading cause of cancer mortality globally, with non-small cell lung cancer (NSCLC) accounting for 85% of cases. Despite advancements in first-line treatments such as immunotherapy and targeted therapies, resistance to these treatments is common, creating a significant unmet need for effective second-line therapies. This review evaluates current and emerging second-line therapeutic options for advanced or metastatic NSCLC, focusing on their efficacy and potential to improve patient outcomes. Anti-angiogenic drugs like ramucirumab combined with chemotherapy, particularly docetaxel, have shown moderate success. Antibody-drug conjugates (ADCs) targeting specific tumor antigens offer a promising avenue for targeted therapy, while chimeric antigen receptor (CAR)-T cell therapy and T-cell receptor therapy leverage the patient's immune system to combat cancer more effectively. mRNA vaccines, although in early stages, show potential for inducing robust immune responses against cancer-specific antigens. Building on this foundation, recent advancements in molecular testing and the exploration of the tumor microenvironment are opening new therapeutic avenues, further enhancing the potential for personalized second-line treatments in NSCLC. While ADCs and bispecific antibodies are gaining traction, more precise biomarkers are needed to optimize treatment response. Regular monitoring through techniques like liquid biopsies allows real-time tracking of mutations such as EGFR T790M, enabling timely therapeutic adjustments. Additionally, the role of neutrophils and macrophages in the tumor microenvironment is increasingly being recognized as a potential therapeutic avenue, with Smad3 emerging as a key target. Further research into drug sequencing, toxicity management, and biomarker development remains crucial to improving NSCLC treatment outcomes.
Collapse
Affiliation(s)
- Kinsley Wang
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Alexis Leyba
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert Hsu
- Department of Medicine, Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
42
|
Fontana P, Du G, Zhang Y, Zhang H, Vora SM, Hu JJ, Shi M, Tufan AB, Healy LB, Xia S, Lee DJ, Li Z, Baldominos P, Ru H, Luo HR, Agudo J, Lieberman J, Wu H. Small-molecule GSDMD agonism in tumors stimulates antitumor immunity without toxicity. Cell 2024; 187:6165-6181.e22. [PMID: 39243763 PMCID: PMC11648675 DOI: 10.1016/j.cell.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/14/2023] [Accepted: 08/06/2024] [Indexed: 09/09/2024]
Abstract
Gasdermin-mediated inflammatory cell death (pyroptosis) can activate protective immunity in immunologically cold tumors. Here, we performed a high-throughput screen for compounds that could activate gasdermin D (GSDMD), which is expressed widely in tumors. We identified 6,7-dichloro-2-methylsulfonyl-3-N-tert-butylaminoquinoxaline (DMB) as a direct and selective GSDMD agonist that activates GSDMD pore formation and pyroptosis without cleaving GSDMD. In mouse tumor models, pulsed and low-level pyroptosis induced by DMB suppresses tumor growth without harming GSDMD-expressing immune cells. Protection is immune-mediated and abrogated in mice lacking lymphocytes. Vaccination with DMB-treated cancer cells protects mice from secondary tumor challenge, indicating that immunogenic cell death is induced. DMB treatment synergizes with anti-PD-1. DMB treatment does not alter circulating proinflammatory cytokine or leukocyte numbers or cause weight loss. Thus, our studies reveal a strategy that relies on a low level of tumor cell pyroptosis to induce antitumor immunity and raise the possibility of exploiting pyroptosis without causing overt toxicity.
Collapse
Affiliation(s)
- Pietro Fontana
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Gang Du
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ying Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Haiwei Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Setu M Vora
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jun Jacob Hu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ming Shi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ahmet B Tufan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Liam B Healy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Shiyu Xia
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Dian-Jang Lee
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Zhouyihan Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Pilar Baldominos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Heng Ru
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA 02115, USA; Department of Laboratory Medicine, Boston Children's Hospital, Enders Research Building, Room 814, Boston, MA 02115, USA
| | - Judith Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Huang H, Tong QS, Chen Y, Liu XY, Liu R, Shen S, Du JZ, Wang J. PAMAM-Based Polymeric Immunogenic Cell Death Inducer To Potentiate Cancer Immunotherapy. J Am Chem Soc 2024; 146:29189-29198. [PMID: 39387453 DOI: 10.1021/jacs.4c11636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Immunogenic cell death (ICD) has been widely employed to potentiate cancer immunotherapy due to its capability to activate the anticancer immune response. Although various ICD inducers have been described, the development of synthetic materials with intrinsic ICD-inducing competency has rarely been reported. Herein, we identify a derivative of the fourth generation polyamidoamine (PAMAM) modified with multiple seven-membered heterocyclic rings, G4P-C7A, as a robust ICD inducer. G4P-C7A evokes characteristic release of damage-associated molecular patterns in tumor cells and induces efficient dendritic cell maturation. Mechanistic studies suggest that G4P-C7A can selectively accumulate in the endoplasmic reticulum and mitochondria to generate reactive oxygen species. G4P-C7A-treated tumor cells can work as potent vaccines to protect against secondary tumor implantation. Either local or systemic injection of G4P-C7A alone can effectively inhibit tumor growth by eliciting robust antitumor immune response. The combination of G4P-C7A with immunotherapeutic antibodies such as anti-PD1 (aPD-1) and anti-CD47 (aCD47) further potentiates the antitumor effect in either CT26 or 4T1 tumor model. This study offers a simple but effective strategy to induce ICD to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Hua Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Qi-Song Tong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Yang Chen
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiao-Yue Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
44
|
Williams CJ, Gray R, Hills RK, Shires M, Zhang L, Zhao Z, Gardner T, Sapanara N, Xu XM, Bai I, Yan D, Muranyi A, Dance S, Aghaei F, Hemmings G, Hale M, Kurkure U, Guetter C, Richman SD, Hutchins G, Seligmann JF, West NP, Singh S, Shanmugam K, Quirke P. Evaluation of CD3 and CD8 T-Cell Immunohistochemistry for Prognostication and Prediction of Benefit From Adjuvant Chemotherapy in Early-Stage Colorectal Cancer Within the QUASAR Trial. J Clin Oncol 2024; 42:3430-3442. [PMID: 39083705 PMCID: PMC11458110 DOI: 10.1200/jco.23.02030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE High densities of tumor infiltrating CD3 and CD8 T-cells are associated with superior prognosis in colorectal cancer (CRC). Their value as predictors of benefit from adjuvant chemotherapy is uncertain. PATIENTS AND METHODS Tumor tissue from 868 patients in the QUASAR trial (adjuvant fluorouracil/folinic acid v observation in stage II/III CRC) was analyzed by CD3 and CD8 immunohistochemistry. Pathologists, assisted by artificial intelligence, calculated CD3 and CD8 cell densities (cells/mm2) in the core tumor (CT) and invasive margin (IM). Participants were randomly partitioned into training and validation sets. The primary outcome was recurrence-free interval (RFI), 2-year RFI for assessment of biomarker-treatment interactions. Maximum-likelihood methods identified optimal high-risk/low-risk group cutpoints in the training set. Prognostic analyses were repeated in the validation set. RESULTS In the training set, the recurrence rate in the high-risk group was twice that in the low-risk group for all measures (CD3-CT: rate ratio [RR], 2.00, P = .0008; CD3-IM: 2.38, P < .00001; CD8-CT: 2.17, P = .0001; CD8-IM: 2.13, P = .0001). This was closely replicated in the validation set (RR, 1.96, 1.79, 1.72, 1.72, respectively). In multivariate analyses, prognostic effects were similar in colon and rectal cancers, and in stage II and III disease. Proportional reductions in recurrence with adjuvant chemotherapy were of similar magnitude in the high- and low-recurrence risk groups. Combining information from CD3-IM and CD3-CT (CD3 Score) generated high-, intermediate-, and low-risk groups with numbers needed to treat (NNTs) to prevent one disease recurrence being 11, 21, and 36, respectively. CONCLUSION Recurrence rates in the high-risk CD3/CD8 groups are twice those in the low-risk groups. Proportional reductions with chemotherapy are similar, allowing NNTs derived in QUASAR to be updated using contemporary, nonrandomized data sets.
Collapse
Affiliation(s)
- Christopher J.M. Williams
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Richard Gray
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- Deceased
| | - Robert K. Hills
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael Shires
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Liping Zhang
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Zuo Zhao
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Tracie Gardner
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Nancy Sapanara
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Xiao-Meng Xu
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Isaac Bai
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Dongyao Yan
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Andrea Muranyi
- Roche Diagnostics Solutions, Research and Development, Oro Valley, AZ
| | - Sarah Dance
- Roche Diagnostics Limited, Medical Affairs, Burgess Hill, West Sussex, United Kingdom
| | - Faranak Aghaei
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Gemma Hemmings
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Michael Hale
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Uday Kurkure
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Christoph Guetter
- Roche Diagnostics Solutions, Imaging and Algorithms, Digital Pathology, Santa Clara, CA
| | - Susan D. Richman
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Gordon Hutchins
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Jenny F. Seligmann
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Nicholas P. West
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| | - Shalini Singh
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Kandavel Shanmugam
- Roche Diagnostics Solutions, Clinical Development and Medical Affairs, Oro Valley, AZ
| | - Philip Quirke
- Division of Pathology and Data Analytics, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
45
|
D'Andrea VD, Magnani CJ, Ernandez J, Bellmunt J, Mossanen M, Clinton TN, Carvalho FLF, Mouw KW. Impact of DNA Repair Deficiency in the Evolving Treatment Landscape of Bladder Cancer. Curr Urol Rep 2024; 26:12. [PMID: 39382743 DOI: 10.1007/s11934-024-01242-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE OF REVIEW This review explores the current landscape of treatments which target the DNA damage response (DDR) in metastatic and muscle-invasive bladder cancer. It emphasizes recent clinical trials which integrate DDR inhibitors with standard chemotherapy and immunotherapy. RECENT FINDINGS Noteworthy findings include the ATLANTIS trial, which demonstrated prolonged progression-free survival (PFS) in DDR biomarker-selected patients using PARP inhibitors as maintenance after standard chemotherapy. Trials such as BAYOU, which combined immunotherapy with PARP inhibition, similarly suggested a potential therapeutic benefit in DDR biomarker-selected patients with bladder cancer. Efforts to develop bladder-sparing treatment regimens based on DDR-associated mutational profiles, such as the RETAIN and HCRN 16-257 trials, have had mixed outcomes to date. There are now ongoing efforts to combine DDR inhibitors with the newest bladder cancer therapies, such as antibody-drug conjugates. This review highlights the most recent advances in targeting DNA repair deficiency in the evolving treatment landscape of bladder cancer.
Collapse
Affiliation(s)
- Vincent D D'Andrea
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Christopher J Magnani
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John Ernandez
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Joaquim Bellmunt
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Matthew Mossanen
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Timothy N Clinton
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Filipe L F Carvalho
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kent W Mouw
- Brigham & Women's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Tan Y, Liu K, Zhu C, Wang S, Wang Y, Xue J, Ning C, Zhang N, Chao J, Zhang L, Long J, Yang X, Zeng D, Zhao L, Zhao H. Efficacy and safety of lenvatinib combined with anti-PD-1 antibodies plus GEMOX chemotherapy as non-first-line systemic therapy in advanced gallbladder cancer. Cancer Immunol Immunother 2024; 73:240. [PMID: 39358463 PMCID: PMC11447189 DOI: 10.1007/s00262-024-03831-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Lenvatinib, programmed cell death 1 (PD-1) antibodies, and gemcitabine and oxaliplatin (GEMOX) chemotherapy have shown significant antitumor activity as first-line therapy against biliary tract cancer. This study evaluated their efficacy and safety as non-first-line therapy in advanced gallbladder cancer (GBC). METHODS Patients with advanced GBC who received lenvatinib combined with anti-PD-1 antibodies and GEMOX chemotherapy as a non-first-line therapy were retrospectively analyzed. The primary endpoints were overall survival (OS) and progression-free survival (PFS), and the secondary endpoints were objective response rate (ORR) and safety. RESULTS A total of 36 patients with advanced GBC were included in this study. The median follow-up time was 11.53 (95% confidence interval (CI): 2.2-20.9) months, and the ORR was 36.1%. The median OS and PFS were 15.1 (95% CI: 3.2-26.9) and 6.1 (95% CI: 4.9-7.2) months, respectively. The disease control rate (DCR) and clinical benefit rate (CBR) were 75% and 61.1%, respectively. Subgroup analysis demonstrated that patients with programmed cell death-ligand 1 (PD-L1) expression had significantly longer PFS and OS than those without PD-L1 expression. Additionally, patients with a neutrophil-lymphocyte ratio (NLR) < 5.57 had a longer OS than those with an NLR ≥ 5.57. All patients experienced adverse events (AEs), with 61.1% experiencing grade 3 or 4 AEs, including myelosuppression (13.9%) and fatigue (13.3%), alanine transaminase or aspartate transaminase levels (8.3%), and diarrhea (8.3%). No grade 5 AEs were reported. CONCLUSION Anti-PD-1 antibodies combined with lenvatinib and GEMOX chemotherapy are effective and well-tolerated as a non-first-line therapy in advanced GBC. PD-L1 expression and baseline NLR may potentially predict treatment efficacy.
Collapse
Affiliation(s)
- Yang Tan
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Kai Liu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Chengpei Zhu
- Department of General Surgery Center, Beijing YouAn Hospital, Clinical Center for Liver Cancer, Capital Medical University, 8 Xitoutiao, Youanmenwai, Fengtai, Beijing, 100069, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yunchao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jingnan Xue
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Cong Ning
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Daobing Zeng
- Department of General Surgery Center, Beijing YouAn Hospital, Clinical Center for Liver Cancer, Capital Medical University, 8 Xitoutiao, Youanmenwai, Fengtai, Beijing, 100069, China.
| | - Lijin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
47
|
Chen Y, Sun Z, Yin J, Ahmad MU, Zhou Z, Feng W, Yang F, Zhou K, Xie J, Bie C, Chen H, Jiang Y. Digital assessment of tertiary lymphoid structures and therapeutic responses in gastric cancer: a multicentric retrospective study. Int J Surg 2024; 110:6732-6747. [PMID: 38884256 PMCID: PMC11486929 DOI: 10.1097/js9.0000000000001834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) are associated with favorable prognosis and enhanced response to anticancer therapy. A digital assessment of TLSs could provide an objective alternative that mitigates variability inherent in manual evaluation. This study aimed to develop and validate a digital gene panel based on biological prior knowledge for assessment of TLSs, and further investigate its associations with survival and multiple anticancer therapies. MATERIALS AND METHODS The present study involved 1704 patients with gastric cancer from seven cancer centers. TLSs were identified morphologically through hematoxylin-and-eosin staining. The authors further developed a digital score based on targeted gene expression profiling to assess TLSs status, recorded as gene signature of tertiary lymphoid structures (gsTLS). For enhanced interpretability, we employed the SHapley Additive exPlanation (SHAP) analysis to elucidate its contribution to the prediction. The authors next evaluated the signature's associations with prognosis, and investigated its predictive accuracy for multiple anticancer therapies, including adjuvant chemotherapy and immunotherapy. RESULTS The gsTLS panel with nine gene features achieved high accuracies in predicting TLSs status in the training, internal, and external validation cohorts (area under the curve, range: 0.729-0.791). In multivariable analysis, gsTLS remained an independent predictor of disease-free and overall survival (hazard ratio, range: 0.346-0.743, all P <0.05) after adjusting for other clinicopathological variables. SHAP analysis highlighted gsTLS as the strongest predictor of TLSs status compared with clinical features. Importantly, patients with high gsTLS (but not those with low gsTLS) exhibited substantial benefits from adjuvant chemotherapy ( P <0.05). Furthermore, the authors found that the objective response rate to antiprogrammed cell death protein 1 (anti-PD-1) immunotherapy was significantly higher in the high-gsTLS group (40.7%) versus the low-gsTLS group (5.6%, P =0.036), and the diagnosis was independent from Epstein-Barr virus, tumor mutation burden, and programmed cell death-ligand 1 (PD-L1) expression. CONCLUSION The gsTLS digital panel enables accurate assessment of TLSs status, and provides information regarding prognosis and responses to multiple therapies for gastric cancer.
Collapse
Affiliation(s)
- Yan Chen
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Zepang Sun
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junmei Yin
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - M. Usman Ahmad
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zixia Zhou
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Wanying Feng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Yang
- Department of Computer Science, Wake Forest University, Winston Salem
| | - Kangneng Zhou
- College of Computer Science, Nankai University, Tianjin, People’s Republic of China
| | - Jingjing Xie
- Graduate Group of Epidemiology, University of California Davis, Davis, California USA
| | - Caiqun Bie
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Hongzhuan Chen
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
48
|
Owens K, Rahman A, Bozic I. Spatiotemporal dynamics of tumor - CAR T-cell interaction following local administration in solid cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610392. [PMID: 39257746 PMCID: PMC11384001 DOI: 10.1101/2024.08.29.610392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The success of chimeric antigen receptor (CAR) T-cell therapy in treating hematologic malignancies has generated widespread interest in translating this technology to solid cancers. However, issues like tumor infiltration, the immunosuppressive tumor microenvironment, and tumor heterogeneity limit its efficacy in the solid tumor setting. Recent experimental and clinical studies propose local administration directly into the tumor or at the tumor site to increase CAR T-cell infiltration and improve treatment outcomes. Characteristics of the types of solid tumors that may be the most receptive to this treatment approach remain unclear. In this work, we develop a spatiotemporal model for CAR T-cell treatment of solid tumors, and use numerical simulations to compare the effect of introducing CAR T cells via intratumoral injection versus intracavitary administration in diverse cancer types. We demonstrate that the model can recapitulate tumor and CAR T-cell data from imaging studies of local administration of CAR T cells in mouse models. Our results suggest that locally administered CAR T cells will be most successful against slowly proliferating, highly diffusive tumors, which have the lowest average tumor cell density. These findings affirm the clinical observation that CAR T cells will not perform equally across different types of solid tumors, and suggest that measuring tumor density may be helpful when considering the feasibility of CAR T-cell therapy and planning dosages for a particular patient. We additionally find that local delivery of CAR T cells can result in deep tumor responses, provided that the initial CAR T-cell dose does not contain a significant fraction of exhausted cells.
Collapse
Affiliation(s)
- Katherine Owens
- Department of Applied Mathematics, University of Washington, Seattle WA
- Fred Hutchinson Cancer Center, Seattle WA
| | - Aminur Rahman
- Fred Hutchinson Cancer Center, Seattle WA
- Artificial Intelligence Institute in Dynamic Systems, University of Washington, Seattle WA
| | - Ivana Bozic
- Department of Applied Mathematics, University of Washington, Seattle WA
- Fred Hutchinson Cancer Center, Seattle WA
| |
Collapse
|
49
|
Kashimura S, Sato M, Inagaki T, Kin M, Manabe R, Kusumoto S, Horiike A, Tsunoda T, Kogo M. Relationship between the combination of platelet count and neutrophil-lymphocyte ratio and prognosis of patients with advanced non-small cell lung cancer treated with immune checkpoint inhibitors plus chemotherapy: A retrospective cohort study. Thorac Cancer 2024; 15:2049-2060. [PMID: 39193939 PMCID: PMC11444929 DOI: 10.1111/1759-7714.15437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND The relationship between the combination of platelet count and neutrophil-lymphocyte ratio (COP-NLR) and prognosis in patients with advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitor (ICI) combination therapy with chemotherapy remains unclear. Thus, we investigated prognostic factors, including the COP-NLR, to identify patients who could benefit from the therapeutic efficacy of ICI combination therapy for advanced NSCLC. Furthermore, we evaluated the relationship between the COP-NLR score during ICI combination therapy and treatment response. METHODS We conducted a retrospective cohort study of 88 patients with NSCLC who initially received ICI combination therapy. The primary outcome was overall survival (OS). The prognostic factors were extracted using the Cox proportional hazards model. The relationship between COP-NLR score at 3 weeks after starting ICI combination therapy and a good response (complete response [CR] and partial response [PR]) to treatment was analyzed using the chi-square test. RESULTS The median OS was 15.7 months. In the multivariable analysis, Eastern Cooperative Oncology Group Performance Status (ECOG PS) 2, distant metastatic sites ≥2, and baseline COP-NLR scores of 1, 2 were extracted as significant poor prognostic factors. The proportion of patients with CR and PR in the 3-week COP-NLR score of 0 group was significantly higher than that in scores of 1, 2 group. CONCLUSIONS Baseline COP-NLR, ECOG PS, and number of distant metastatic sites were prognostic factors in patients with NSCLC with ICI combination therapy. A lower 3-week COP-NLR was associated with a good response to treatment.
Collapse
Affiliation(s)
- Saeko Kashimura
- Division of Pharmacotherapeutics, Department of Clinical PharmacyShowa University School of PharmacyTokyoJapan
| | - Miki Sato
- Division of Pharmacotherapeutics, Department of Clinical PharmacyShowa University School of PharmacyTokyoJapan
| | - Takahito Inagaki
- Department of Hospital Pharmaceutics, School of PharmacyShowa UniversityTokyoJapan
- Department of PharmacyShowa University Northern Yokohama HospitalKanagawaJapan
| | - Masaoki Kin
- Department of PharmacyShowa University HospitalTokyoJapan
| | - Ryo Manabe
- Division of Respirology and Allergology, Department of MedicineShowa University School of MedicineTokyoJapan
| | - Sojiro Kusumoto
- Division of Respirology and Allergology, Department of MedicineShowa University School of MedicineTokyoJapan
| | - Atsushi Horiike
- Division of Medical Oncology, Department of MedicineShowa University School of MedicineTokyoJapan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of MedicineShowa University School of MedicineTokyoJapan
| | - Mari Kogo
- Division of Pharmacotherapeutics, Department of Clinical PharmacyShowa University School of PharmacyTokyoJapan
| |
Collapse
|
50
|
Sriramulu S, Thoidingjam S, Speers C, Nyati S. Present and Future of Immunotherapy for Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3250. [PMID: 39409871 PMCID: PMC11475478 DOI: 10.3390/cancers16193250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Triple-negative breast cancer (TNBC) lacks the expression of estrogen receptors (ERs), human epidermal growth factor receptor 2 (HER2), and progesterone receptors (PRs). TNBC has the poorest prognosis among breast cancer subtypes and is more likely to respond to immunotherapy due to its higher expression of PD-L1 and a greater percentage of tumor-infiltrating lymphocytes. Immunotherapy has revolutionized TNBC treatment, especially with the FDA's approval of pembrolizumab (Keytruda) combined with chemotherapy for advanced cases, opening new avenues for treating this deadly disease. Although immunotherapy can significantly improve patient outcomes in a subset of patients, achieving the desired response rate for all remains an unmet clinical goal. Strategies that enhance responses to immune checkpoint blockade, including combining immunotherapy with chemotherapy, molecularly targeted therapy, or radiotherapy, may improve response rates and clinical outcomes. In this review, we provide a short background on TNBC and immunotherapy and explore the different types of immunotherapy strategies that are currently being evaluated in TNBC. Additionally, we review why combination strategies may be beneficial, provide an overview of the combination strategies, and discuss the novel immunotherapeutic opportunities that may be approved in the near future for TNBC.
Collapse
Affiliation(s)
- Sushmitha Sriramulu
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Shivani Thoidingjam
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiation Oncology, UH Seidman Cancer Center, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shyam Nyati
- Department of Radiation Oncology, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI 48202, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|